Body Mass Index in Multiple Sclerosis modulates ceramide-induced DNA methylation and disease course

Kamilah Castro, Achilles Ntranos, Mario Amatruda, Maria Petracca, Peter Kosa, Emily Y Chen, Johannes Morstein, Dirk Trauner, Corey T Watson, Michael A Kiebish, Bibiana Bielekova, Matilde Inglese, Ilana Katz Sand, Patrizia Casaccia, Kamilah Castro, Achilles Ntranos, Mario Amatruda, Maria Petracca, Peter Kosa, Emily Y Chen, Johannes Morstein, Dirk Trauner, Corey T Watson, Michael A Kiebish, Bibiana Bielekova, Matilde Inglese, Ilana Katz Sand, Patrizia Casaccia

Abstract

Background: Multiple Sclerosis (MS) results from genetic predisposition and environmental variables, including elevated Body Mass Index (BMI) in early life. This study addresses the effect of BMI on the epigenome of monocytes and disease course in MS.

Methods: Fifty-four therapy-naive Relapsing Remitting (RR) MS patients with high and normal BMI received clinical and MRI evaluation. Blood samples were immunophenotyped, and processed for unbiased plasma lipidomic profiling and genome-wide DNA methylation analysis of circulating monocytes. The main findings at baseline were validated in an independent cohort of 91 therapy-naïve RRMS patients. Disease course was evaluated by a two-year longitudinal follow up and mechanistic hypotheses tested in human cell cultures and in animal models of MS.

Findings: Higher monocytic counts and plasma ceramides, and hypermethylation of genes involved in negative regulation of cell proliferation were detected in the high BMI group of MS patients compared to normal BMI. Ceramide treatment of monocytic cell cultures increased proliferation in a dose-dependent manner and was prevented by DNA methylation inhibitors. The high BMI group of MS patients showed a negative correlation between monocytic counts and brain volume. Those subjects at a two-year follow-up showed increased T1 lesion load, increased disease activity, and worsened clinical disability. Lastly, the relationship between body weight, monocytic infiltration, DNA methylation and disease course was validated in mouse models of MS.

Interpretation: High BMI negatively impacts disease course in Multiple Sclerosis by modulating monocyte cell number through ceramide-induced DNA methylation of anti-proliferative genes. FUND: This work was supported by funds from the Friedman Brain Institute, NIH, and Multiple Sclerosis Society.

Keywords: Epigenetics; Immunity; Lipids; Neurodegeneration; Obesity.

Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Immune profiling reveals a BMI-dependent increase in blood monocyte counts in MS patients but not healthy individuals. Immune cell populations from fresh whole blood were quantified using flow cytometry. (a) Differences in T cell counts due to BMI were assessed in a primary cohort of MS patients (n = 52). (b) Differences in monocyte counts due to BMI were assessed in a primary cohort of MS patients (n = 52), a validation cohort of MS patients (n = 91), and a cohort of healthy individuals (n = 50). One-way MANCOVA was used to adjust for age and sex, followed by pair-wise comparisons with Bonferroni correction to determine statistically significant differences in the two BMI groups (normal BMI = white dots, high BMI = gray dots) (*p < .05).
Fig. 2
Fig. 2
Lipidomic profiling reveals increased abundance of ceramide species in MS patients with high BMI, but not in healthy individuals. Unbiased lipidomic analysis was performed on plasma samples from MS patients (n = 48) and healthy individuals (n = 40) using an MS/MSALL platform. Lipids that differed in abundance due to BMI were assessed using multiple t-tests with 5% FDR correction, q < 0.05 were considered significant (*q < 0.05, **q < 0.01). (a) Pie chart representation of numbers and percentage of lipids in MS patients by lipid family and class. Red = increased abundance, blue = decreased abundance, white = no change in patients with high BMI. (b) Relative abundance of ceramide species with 18:1 backbone that were significantly different in abundance in MS patients with high BMI (gray dots) compared to those with normal BMI (white). (c) The most highly significantly abundant ceramide species in MS patients were not statistically different in healthy individuals with high BMI (gray dots) compared to those with normal BMI (white). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 3
Fig. 3
Exposure to ceramides results in dose-dependent increase in proliferation and DNA methylation. (a) Cultured monocytes (i.e. THP1 cells) were treated with fluorescently labeled Ceramide C16 (NBD-Ceramide C16) or untreated for 24 h. 3D reconstruction of the cells was conducted on Imaris to visualize localization of NBD-Ceramide (green). (b) Cultures either untreated or treated with Ceramide C16 (Cer C16) for 1, 12, or 24 h were fixed at the same time. After immunocytochemistry for Ki67 to detect proliferating cells or5-methylcytosine as marker for global DNA methylation the percentage of cells relative to the total DAPI+ nuclei was calculated and plotted in the graphs. Statistical differences in the percentage of Ki67+ cells and percentage of 5mc + cells relative to total DAPI+ nuclei were assessed by Student's t-test. (c) Cultures were treated for 1 (left panels) or 24 (right panels) hours with concentrations of a mixture of ceramides C16, C22, and C24:1, mimicking those detected in the plasma of MS patients with normal (Low) or high BMI (High) in the absence or presence of the DNA methylation inhibitor, 5-aza-2′-deoxycytidine (5-aza). After fixation, cells were stained for Ki67 (green) as proliferation marker, 5-methylcytosine (5mC, red), as marker for global DNA methylation and DAPI (blue), to identify nuclei. Confocal images were acquired at 20× magnification using the Zeiss LSM800 confocal (scale bar = 25um). High and High+5-aza groups were compared to Low Cer at 24 h using Dunnett's multiple comparisons test (*p p < .01) (n = 3 independent biological replicates, each conducted in triplicate wells and analyzing 3 images/well). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 4
Fig. 4
Genome-wide distribution of DNA methylation marks in monocytes of high BMI MS patients. DNA methylation analysis was conducted on DNA from monocytes of MS patients using the Infinium HumanMethylation450 BeadChip. DNA samples from the monocytes of MS patients with high BMI (n = 23) were compared to those from patients with normal BMI (n = 25). Differentially methylated regions (DMRs) were identified using a 1 kb sliding window and significantly different CpGs were determined by evaluating the combined p values of CpG sites within each region, using the Stouffer's method with a 1% FDR correction. (a) Methylation values of individual CpGs within DMRs were measured as beta values, or the signal intensity from the methylated probe in relation to the sum of intensities from both methylated and unmethylated probes. Beta values of CpGs in MS samples with high BMI were subtracted from those in with normal BMI and a frequency histogram of the beta value differences was plotted to visualize the frequency of beta value differences in a given range. Positive beta value differences, indicating DNA hypermethylation in high BMI MS samples, are plotted in red, whereas negative differences, indicating DNA hypomethylation, are in blue. (b) Genomic distribution of the hypermethylated DMR in patients with high BMI (black bars), compared to the distribution of all the CpGs in the array (white bar). The relative proportions of CpGs in islands, shores (2 kb flanking the islands), shelves (2 kb flanking the shores) and sea (regions outside the previous three categories) are indicated. (c) Distribution of the hypermethylated CpGs distributed within in MS patients with high BMI (black bars) within distinct gene features, compared to the distribution of all the CpGs in the array (white bar). The relative proportion of differentially methylated CpGs in high BMI MS patients is represented relative to RefSeq gene promoters (2 kb from TSS), 5’UTR, 1st exon, gene bodies, 3’UTR and intergenic regions. Note that the CpGs hypermethylated in MS patients with high BMI were enriched at promoters and intergenic region and depleted at gene bodies, consistent with a repressive pattern of DNA methylation. Enrichment defined as statistically significantly different from the proportion of total array CpGs in a given location, which was calculated by chi- squared test, *p < .05, **p < .01. (d) Pathway enrichment analysis was performed on hypermethylated CpGs using the KEGG database on Enrichr. Enrichment score was calculated as the -log (adjusted p value). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 5
Fig. 5
Chromosomal distribution and identification of genes with most significantly hypermethylated regions in MS patients with high BMI identifies negative regulators of cell proliferation. (a) Circos plot representing the chromosomal distribution of DMRs and CpGs within DMRs. Chromosomal numbers are indicated in the innermost circle. The inner red and blue circle depict the beta value differences for each CpG within a DMR. The more hypermethylated the CpG (bright red) the farther it is from the center of the circos plot, and the more hypomethylated the CpG (blue) the closer it is to the center of the plot. Dark red lines in the outer circle denote the genomic location of the DMRs and dark red dots indicate the mean beta value difference for each DMR. Genes overlapping DMRs are indicated in the outermost circle and genes with the greatest methylation differences are highlighted in red. (b) Manhattan plot highlighting the overall methylation difference, or mean beta change, for each DMR on each chromosome. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 6
Fig. 6
Exposure to ceramides results in increased DNA methylation and reduced transcription of negative regulators of cell proliferation. (a) Cultured human monocytes (i.e. THP1 cells) were either untreated (blue) or treated with Ceramide C16 (red) for 24 h. DNA was harvested and processed for Sequenom Mass Array EpiTyper, in order to quantify differential methylation of CpGs (Y axis) at the indicated genomic locations (X axis). The gene name is shown on top of each graph. (b) samples from the same cells described in (a) were used for RNA isolation and quantification of the corresponding gene transcripts by using real time-PCR and normalized to reference gene transcripts. (c) Cultures were treated for 24 h with concentrations of a mixture of ceramides C16, C22, and C24:1, mimicking those detected in the plasma of MS patients with normal (Low) or high BMI (High) in the absence or presence of the DNA methylation inhibitor, 5-aza-2′-deoxycytidine (5-aza). DNA was harvested and processed for Sequenom Mass Array EpiTyper, in order to quantify differential methylation of CpGs (Y axis) at the indicated genomic locations (X axis). The gene name is shown on top of each graph. Overall methylation differences for the entire region of interest for each gene were assessed using two-way ANOVA (*p < .05, ***p < .0001) (n = 3/group, n = 2 replicates/group). (d) Transcript levels of NRXN1, FZD7, and TP63 were assessed using real time-PCR and normalized to the levels of thehousekeeping gene, RPLP0. Cer C16 groups were compared to Untreated groups using Student's t-test, while the High and High +5-aza groups were compared to the Low group, using Dunnett's multiple comparisons test (*p < .05, **p < .01, ***p < .001) (n = 3 independent experiments, n = 3 biological replicates/experiment, n = 3 technical replicates/biological replicate). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 7
Fig. 7
Monocytes uniquely correlate with reduced brain volume in overweight/obese MS patients, who exhibit worsened disease course at the two-year follow up. (a) Brain volumetric analysis was performed on T1-weighted images from MRI scans and correlated with CD14+ monocyte counts from fresh whole blood, which had been obtained by flow cytometry, in a primary MS cohort (n = 41), validation MS cohort (n = 80) and a healthy cohort (n = 48) using linear regression, p < .05 considered significant. (b) Differences in the proportion of patients who showed changes in T1 lesion load (ΔT1) <1 ml, between 0 and 1 ml, and >1 ml during a 2 year period, between MS patients with normal or high BMI were assessed by chi-squared test (*p < .05). (c) Differences in the proportion of patients who had disease activity, defined by clinical relapse or new lesions, during a 2 year period, between MS patients with normal or high BMI were assessed by chi-squared test (*p < .05). (d) Change in EDSS in a 2 year period was categorized as better (<−0.5), same (−0.5 to 0.5), or worse (>0.5) and correlated with BMI using ordinal regression, p < .05 considered significant.
Fig. 8
Fig. 8
High-fat diet fed mice show increased monocytic infiltration and greater neurological deficits in an experimental model of MS. (a) Female C57/bl6 mice were fed either a low-fat (LFD) or high-fat diet (HFD) for 5 weeks, and then immunized with 200 ng MOG35–55 using a protocol to induce EAE. (b) Ascending paralysis was graded on a scale of 0 to 5 with high scores indicating increased clinical disability (black = LFD-EAE mice, gray = HFD-EAE mice). Differences in EAE scores were evaluated between LFD-EAE and HFD-EAE mice at each time point using Mann-Whitney U test, respectively (*p < .05, **p < .01) (n = 15/group). (c) Weight change from baseline (i.e. day of immunization) was calculated for each time point and differences between LFD-EAE and HFD-EAE mice were evaluated by Sidak's multiple comparisons test (**p < .01). (d) Area under the curve of the EAE disease course and maximum EAE score were also used to evaluate differences in disease course and statistical analysis was performed using Mann-Whitney U test (*p < .05). (e-g) Mice were sacrificed at the peak of disease and spinal cord tissue sectioned for histological analysis. Spinal cord sections were stained with Fluoromyelin (FM)to stain myelin and lesion areas and Neurofilament H (NFH) to stain axons. Images were acquired using the Zeiss LSM 800 confocal microscope and the 20× objective (scale bar = 50um). Myelin and axonal content were quantified as the area of FM or NFH positivity, respectively, in relation to the whole spinal cord area. Spinal cord sections were also stained with Iba1 and CD45 to mark peripheral monocytes/activated microglia (Iba1+ CD45+) and leukocytes (CD45+) and counterstained with DAPI, to identify nuclei. Confocal images were acquired using the Zeiss LSM 800 and the 20× objective (scale bar = 25um). Iba1 + CD45+ area and CD45+ area was quantified within the lesion to enhance representation of peripheral monocytes and peripheral leukocytes, respectively, and was calculated as a percentage in relation to the total spinal cord area. Spinal cord sections were stained with 5mc and Iba1 to assess DNA methylation levels in monocytes. Images were acquired using the Zeiss LSM 800 at 63× (scale bar = 10um). 5mc intensity was normalized to DAPI intensity in Iba1+ cells within the lesion. Statistical differences between LFD-EAE (white) and HFD-EAE (gray) groups were assessed by Student's t-test (*p < .05) (n = 3 mice/group, n = 2 sections/mouse, n = 3 images/section).

References

    1. Ogden C.L., Carroll M.D., Kit B.K., Flegal K.M. Prevalence of childhood and adult obesity in the United States, 2011-2012. JAMA. 2014;311:806–814.
    1. Aune D., Sen A., Prasad M., Norat T., Janszky I., Tonstad S. BMI and all cause mortality: systematic review and non-linear dose-response meta-analysis of 230 cohort studies with 3.74 million deaths among 30.3 million participants. BMJ. 2016;353
    1. Masters R.K., Reither E.N., Powers D.A., Yang Y.C., Burger A., Link B.G. The impact of obesity on US mortality levels: the importance of age and cohort factors in population estimates. Am J Public Health. 2013;103:1895–1901.
    1. Peitz G.W., Troyer J., Jones A.E., Shapiro N.I., Nelson R.D., Hernandez J. Association of body mass index with increased cost of care and length of stay for emergency department patients with chest pain and dyspnea. Circ Cardiovasc Qual Outcomes. 2014;7:292–298.
    1. Teuner C.M., Menn P., Heier M., Holle R., John J., Wolfenstetter S.B. Impact of BMI and BMI change on future drug expenditures in adults: results from the MONICA/KORA cohort study. BMC Health Serv Res. 2013;13
    1. Dee A., Kearns K., O'Neill C., Sharp L., Staines A., O'Dwyer V. The direct and indirect costs of both overweight and obesity: a systematic review. BMC Res Notes. 2014;7
    1. Beydoun M.A., Beydoun H., Wang Y. Obesity and central obesity as risk factors for incident dementia and its sub-types: a systematic review and meta-analysis. Obes Rev Off J Int Assoc Study Obes. 2008;9:204–218.
    1. Ogrodnik M., Zhu Y., Langhi L.G.P., Tchkonia T., Krüger P., Fielder E. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab. 2019
    1. Whitmer R.A., Gustafson D.R., Barrett-Connor E., Haan M.N., Gunderson E.P., Yaffe K. Central obesity and increased risk of dementia more than three decades later. Neurology. 2008;71:1057.
    1. Abbott R.D., Ross G.W., White L.R., Nelson J.S., Masaki K.H., Tanner C.M. Midlife adiposity and the future risk of Parkinson's disease. Neurology. 2002;59:1051.
    1. Hu G., Jousilahti P., Nissinen A., Antikainen R., Kivipelto M., Tuomilehto J. Body mass index and the risk of Parkinson disease. Neurology. 2006;67:1955.
    1. Hedström A.K., Olsson T., Alfredsson L. High body mass index before age 20 is associated with increased risk for multiple sclerosis in both men and women. Mult Scler J. 2012;18:1334–1336.
    1. Munger K.L., Chitnis T., Ascherio A. Body size and risk of MS in two cohorts of US women. Neurology. 2009;73:1543–1550.
    1. Wesnes K., Riise T., Casetta I., Drulovic J., Granieri E., Holmøy T. Body size and the risk of multiple sclerosis in Norway and Italy: the EnvIMS study. Mult Scler J. 2015;21:388–395.
    1. Frohman E.M., Racke M.K., Raine C.S. Multiple sclerosis—the plaque and its pathogenesis. N Engl J Med. 2006;354:942–955.
    1. Hedström A.K., Olsson T., Alfredsson L. Body mass index during adolescence, rather than childhood, is critical in determining MS risk. Mult Scler J. 2016;22:878–883.
    1. Langer-Gould A., Brara S.M., Beaber B.E., Koebnick C. Childhood obesity and risk of pediatric multiple sclerosis and clinically isolated syndrome. Neurology. 2013;80:548–552.
    1. Munger K.L., Bentzen J., Laursen B., Stenager E., Koch-Henriksen N., Sørensen T.I. Childhood body mass index and multiple sclerosis risk: a long-term cohort study. Mult Scler J. 2013;19:1323–1329.
    1. Chitnis T., Graves J., Weinstock-Guttman B., Belman A., Olsen C., Misra M. Distinct effects of obesity and puberty on risk and age at onset of pediatric MS. Ann Clin Transl Neurol. 2016;3:897–907.
    1. Gianfrancesco M.A., Acuna B., Shen L., Briggs F.B., Quach H., Bellesis K.H. Obesity during childhood and adolescence increases susceptibility to multiple sclerosis after accounting for established genetic and environmental risk factors. Obes Res Clin Pract. 2014;8:e435–e447.
    1. Bove R., Musallam A., Xia Z., Baruch N., Messina S., Healy B.C. Longitudinal BMI trajectories in multiple sclerosis: sex differences in association with disease severity. Mult Scler Relat Disord. 2016;8:136–140.
    1. Kappus N., Weinstock-Guttman B., Hagemeier J., Kennedy C., Melia R., Carl E. Cardiovascular risk factors are associated with increased lesion burden and brain atrophy in multiple sclerosis. J Neurol Neurosurg Psychiatry. 2015;87:181–187. [jnnp–2014].
    1. Mowry E.M., Azevedo C.J., McCulloch C.E., Okuda D.T., Lincoln R.R., Waubant E. Body mass index, but not vitamin D status, is associated with brain volume change in MS. Neurology. 2018;91:e2256–e2264.
    1. Oliveira S.R., Simão A.N.C., Kallaur A.P., de Almeida E.R.D., Morimoto H.K., Lopes J. Disability in patients with multiple sclerosis: influence of insulin resistance, adiposity, and oxidative stress. Nutrition. 2014;30:268–273.
    1. Tettey P., Simpson S., Taylor B.V., van der Mei I.A. Vascular comorbidities in the onset and progression of multiple sclerosis. J Neurol Sci. 2014;347:23–33.
    1. Manouchehrinia A., Hedström A.K., Alfredsson L., Olsson T., Hillert J., Ramanujam R. Association of pre-Disease Body Mass Index with Multiple Sclerosis Prognosis. Front Neurol. 2018;9
    1. Paz-Ballesteros WC, Monterrubio-Flores EA, de Jesús Flores-Rivera J, Corona-Vázquez T, Hernández-Girón C. Cigarette smoking, alcohol consumption and overweight in multiple sclerosis: disability progression. Arch Med Res 2017;48:113–20. doi:10.1016/j.arcmed.2017.03.002.
    1. Tonks K.T., Coster A.C., Christopher M.J., Chaudhuri R., Xu A., Gagnon-Bartsch J. Skeletal muscle and plasma lipidomic signatures of insulin resistance and overweight/obesity in humans. Obesity. 2016;24:908–916.
    1. Goodpaster B.H., Wolf D. Skeletal muscle lipid accumulation in obesity, insulin resistance, and type 2 diabetes. Pediatr Diabetes. 2004;5:219–226.
    1. Hla T., Dannenberg A.J. Sphingolipid signaling in metabolic disorders. Cell Metab. 2012;16:420–434.
    1. Cowart L.A. Sphingolipids: players in the pathology of metabolic disease. Trends Endocrinol Metab. 2009;20:34–42.
    1. Wymann M.P., Schneiter R. Lipid signalling in disease. Nat Rev Mol Cell Biol. 2008;9:162–176.
    1. Halmer R., Walter S. Fa\s sbender K. Sphingolipids: important players in multiple sclerosis. Cell Physiol Biochem. 2014;34:111–118.
    1. Cumings J.N., Goodwin H. Sphingolipids and phospholipids of myelin in multiple sclerosis. The Lancet. 1968;292:664–665.
    1. Wheeler D., Bandaru V.V.R., Calabresi P.A., Nath A., Haughey N.J. A defect of sphingolipid metabolism modifies the properties of normal appearing white matter in multiple sclerosis. Brain. 2008;131:3092–3102.
    1. Vidaurre O.G., Haines J.D., Sand I.K., Adula K.P., Huynh J.L., McGraw C.A. Cerebrospinal fluid ceramides from patients with multiple sclerosis impair neuronal bioenergetics. Brain. 2014;137:2271–2286. awu139.
    1. Kurz J., Brunkhorst R., Foerch C., Blum L., Henke M., Gabriel L. The relevance of ceramides and their synthesizing enzymes for multiple sclerosis. Clin Sci. 2018 CS20180506.
    1. Kurz J., Parnham M.J., Geisslinger G., Schiffmann S. Ceramides as novel disease biomarkers. Trends Mol Med. 2019;25:20–32.
    1. Summers S.A. Could ceramides become the new cholesterol? Cell Metab. 2018;27:276–280.
    1. Hla T., Kolesnick R. C16: 0-ceramide signals insulin resistance. Cell Metab. 2014;20:703–705.
    1. Polman C.H., Reingold S.C., Banwell B., Clanet M., Cohen J.A., Filippi M. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol. 2011;69:292–302.
    1. Kurtzke J.F. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS) Neurology. 1983;33:1444–1452.
    1. Han S., Lin Y.C., Wu T., Salgado A.D., Mexhitaj I., Wuest S.C. Comprehensive immunophenotyping of cerebrospinal fluid cells in patients with neuroimmunological diseases. J Immunol Baltim Md. 2014;1950(192):2551–2563.
    1. Bielekova B., Catalfamo M., Reichert-Scrivner S., Packer A., Cerna M., Waldmann T.A. Regulatory CD56bright natural killer cells mediate immunomodulatory effects of IL-2Rα-targeted therapy (daclizumab) in multiple sclerosis. Proc Natl Acad Sci. 2006;103:5941–5946.
    1. Gao F., McDaniel J., Chen E.Y., Rockwell H., Lynes M.D., Tseng Y.-H. Monoacylglycerol analysis using MS/MS(ALL) quadruple time of flight mass spectrometry. Metabolites. 2016;6
    1. Han X., Gross R.W. Shotgun lipidomics: electrospray ionization mass spectrometric analysis and quantitation of cellular lipidomes directly from crude extracts of biological samples. Mass Spectrom Rev. 2005;24:367–412.
    1. Simons B., Kauhanen D., Sylvanne T., Tarasov K., Duchoslav E., Ekroos K. Shotgun Lipidomics by sequential precursor ion fragmentation on a hybrid quadrupole time-of-flight mass spectrometer. Metabolites. 2012;2:195–213.
    1. Du P., Zhang X., Huang C.-C., Jafari N., Kibbe W.A., Hou L. Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics. 2010;11
    1. Zhou W., Laird P.W., Shen H. Comprehensive characterization, annotation and innovative use of Infinium DNA methylation BeadChip probes. Nucleic Acids Res. 2017;45:e22.
    1. Nordlund J., Bäcklin C.L., Wahlberg P., Busche S., Berglund E.C., Eloranta M.L. Genome-wide signatures of differential DNA methylation in pediatric acute lymphoblastic leukemia. Genome Biol. 2013;14:r105.
    1. Tian Y., Morris T.J., Webster A.P., Yang Z., Beck S., Feber A. ChAMP: updated methylation analysis pipeline for Illumina BeadChips. Bioinformatics. 2017;33:3982–3984.
    1. Huynh J.L., Garg P., Thin T.H., Yoo S., Dutta R., Trapp B.D. Epigenome-wide differences in pathology-free regions of multiple sclerosis-affected brains. Nat Neurosci. 2014;17:121–130.
    1. Watson C.T., Roussos P., Garg P., Ho D.J., Azam N., Katsel P.L. Genome-wide DNA methylation profiling in the superior temporal gyrus reveals epigenetic signatures associated with Alzheimer's disease. Genome Med. 2016;8
    1. Ritchie M.E., Phipson B., Wu D., Hu Y., Law C.W., Shi W. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.
    1. Peters T.J., Buckley M.J., Statham A.L., Pidsley R., Samaras K., V Lord R. De novo identification of differentially methylated regions in the human genome. Epigenetics Chromatin. 2015;8
    1. Whitlock M.C. Combining probability from independent tests: the weighted Z-method is superior to Fisher's approach. J Evol Biol. 2005;18:1368–1373.
    1. Storey J.D., Tibshirani R. Statistical significance for genomewide studies. Proc Natl Acad Sci. 2003;100:9440–9445.
    1. Kuleshov M.V., Jones M.R., Rouillard A.D., Fernandez N.F., Duan Q., Wang Z. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44:W90–W97.
    1. Chen E.Y., Tan C.M., Kou Y., Duan Q., Wang Z., Meirelles G.V. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinforma. 2013;14:128.
    1. Smith S.M., Zhang Y., Jenkinson M., Chen J., Matthews P.M., Federico A. Accurate, robust, and automated longitudinal and cross-sectional brain change analysis. NeuroImage. 2002;17:479–489.
    1. Shiee N., Bazin P.-L., Ozturk A., Reich D.S., Calabresi P.A., Pham D.L. A topology-preserving approach to the segmentation of brain images with multiple sclerosis lesions. NeuroImage. 2010;49:1524–1535.
    1. Ito S., D'Alessio A.C., Taranova O.V., Hong K., Sowers L.C., Zhang Y. Role of Tet proteins in 5mC to 5hmC conversion, ES cell self-renewal, and ICM specification. Nature. 2010;466:1129–1133.
    1. Haines J.D., Vidaurre O.G., Zhang F., Riffo-Campos Á.L., Castillo J., Casanova B. Multiple sclerosis patient-derived CSF induces transcriptional changes in proliferating oligodendrocyte progenitors. Mult Scler Houndmills Basingstoke Engl. 2015;21:1655–1669.
    1. Miller S.D., Karpus W.J., Davidson T.S. Experimental autoimmune encephalomyelitis in the mouse. Curr Protoc Immunol Ed John E Coligan Al. 2007 CHAPTER:Unit-15.1.
    1. Pino P.A., Cardona A.E. Isolation of brain and spinal cord mononuclear cells using Percoll gradients. J Vis Exp JoVE. 2011
    1. Barrès R., Osler M.E., Yan J., Rune A., Fritz T., Caidahl K. Non-CpG methylation of the PGC-1α promoter through DNMT3B controls mitochondrial density. Cell Metab. 2009;10:189–198.
    1. Dekkers K.F., van Iterson M., Slieker R.C., Moed M.H., Bonder M.J., van Galen M. Blood lipids influence DNA methylation in circulating cells. Genome Biol. 2016;17
    1. Flores-Sierra J., Arredondo-Guerrero M., Cervantes-Paz B., Rodríguez-Ríos D., Alvarado-Caudillo Y., Nielsen F.C. The trans fatty acid elaidate affects the global DNA methylation profile of cultured cells and in vivo. Lipids Health Dis. 2016;15
    1. de la Rocha C., Pérez-Mojica J.E., León S.Z.-D., Cervantes-Paz B., Tristán-Flores F.E., Rodríguez-Ríos D. Associations between whole peripheral blood fatty acids and DNA methylation in humans. 2016;6
    1. Silva-Martinez G.A., Rodriguez-Rios D., Alvarado-Caudillo Y., Vaquero A., Esteller M., Carmona F.J. Arachidonic and oleic acid exert distinct effects on the DNA methylome. Epigenetics. 2016;11:321–334.
    1. Hammad S.M., Pierce J.S., Soodavar F., Smith K.J., Gadban M.M.A., Rembiesa B. Blood sphingolipidomics in healthy humans: impact of sample collection methodology. J Lipid Res. 2010;51:3074–3087.
    1. Ottenlinger F.M., Mayer C.A., Ferreirós N., Schreiber Y., Schwiebs A., Schmidt K.G. Interferon-Beta increases plasma ceramides of specific chain length in multiple sclerosis patients, unlike Fingolimod or Natalizumab. Front Pharmacol. 2016;7
    1. Bock C. Analysing and interpreting DNA methylation data. Nat Rev Genet. 2012;13:705–719.
    1. Kinzfogl J., Hangoc G., Broxmeyer H.E. Neurexophilin 1 suppresses the proliferation of hematopoietic progenitor cells. Blood. 2011;118:565–575.
    1. Manni I., Artuso S., Careccia S., Rizzo M.G., Baserga R., Piaggio G. The microRNA miR-92 increases proliferation of myeloid cells and by targeting p63 modulates the abundance of its isoforms. FASEB J. 2009;23:3957–3966.
    1. Chakrabarti R., Wei Y., Hwang J., Hang X., Andres Blanco M., Choudhury A. ΔNp63 promotes stem cell activity in mammary gland development and basal-like breast cancer by enhancing Fzd7 expression and Wnt signalling. Nat Cell Biol. 2014;16:1004–1015.
    1. Bermel R.A., Bakshi R. The measurement and clinical relevance of brain atrophy in multiple sclerosis. Lancet Neurol. 2006;5:158–170.
    1. Fisher E., Rudick R.A., Cutter G., Baier M., Miller D., Weinstock-Guttman B. Relationship between brain atrophy and disability: an 8-year follow-up study of multiple sclerosis patients. Mult Scler J. 2000;6:373–377.
    1. Popescu V., Agosta F., Hulst H.E., Sluimer I.C., Knol D.L., Sormani M.P. Brain atrophy and lesion load predict long term disability in multiple sclerosis. J Neurol Neurosurg Psychiatry. 2013;84:1082–1091.
    1. Giorgio A., Stromillo M.L., Bartolozzi M.L., Rossi F., Battaglini M., De Leucio A. Relevance of hypointense brain MRI lesions for long-term worsening of clinical disability in relapsing multiple sclerosis. Mult Scler J. 2014;20:214–219.
    1. Radue E.-W., Barkhof F., Kappos L., Sprenger T., Häring D.A., de Vera A. Correlation between brain volume loss and clinical and MRI outcomes in multiple sclerosis. Neurology. 2015;84:784–793.
    1. Sailer M., Losseff N.A., Wang L., Gawne-Cain M.L., Thompson A.J., Miller D.H. T1 lesion load and cerebral atrophy as a marker for clinical progression in patients with multiple sclerosis. A prospective 18 months follow-up study. Eur J Neurol. 2001;8:37–42.
    1. Simon J.H., Jacobs L., Simonian N. T1-Hypointense lesions (T1 black holes) in mild-to-moderate disability relapsing multiple sclerosis. In: Hommes O.R., Comi G., editors. Early Indic Early Treat Neuroprotection Mult Scler. Springer Milan; Milano: 2004. pp. 135–139.
    1. Christine Poitou, Elise Dalmas, Mariana Renovato, Vanessa Benhamo, Froogh Hajduch, Meriem Abdennour. CD14dimCD16+ and CD14+CD16+ monocytes in obesity and during weight loss. Arterioscler Thromb Vasc Biol. 2011;31:2322–2330.
    1. Devevre E.F., Renovato-Martins M., Clement K., Sautes-Fridman C., Cremer I., Poitou C. Profiling of the three circulating monocyte subpopulations in human obesity. J Immunol Baltim Md. 2015;1950(194):3917–3923.
    1. Wouters K., Gaens K., Bijnen M., Verboven K., Jocken J., Wetzels S. Circulating classical monocytes are associated with CD11c+ macrophages in human visceral adipose tissue. Sci Rep. 2017;7
    1. Hyson D.A., Paglieroni T.G., Wun T., Rutledge J.C. Postprandial Lipemia is associated with platelet and monocyte activation and increased monocyte cytokine expression in Normolipemic men. Clin Appl Thromb. 2002;8:147–155.
    1. Haus J.M., Kashyap S.R., Kasumov T., Zhang R., Kelly K.R., Defronzo R.A. Plasma ceramides are elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance. Diabetes. 2009;58:337–343.
    1. Hilvo M., Salonurmi T., Havulinna A.S., Kauhanen D., Pedersen E.R., Tell G.S. Ceramide stearic to palmitic acid ratio predicts incident diabetes. Diabetologia. 2018;61:1424–1434.
    1. Lopez X., Goldfine A.B., Holland W.L., Gordillo R., Scherer P.E. Plasma ceramides are elevated in female children and adolescents with type 2 diabetes. J Pediatr Endocrinol Metab JPEM. 2013;26:995–998.
    1. Wigger L., Cruciani-Guglielmacci C., Nicolas A., Denom J., Fernandez N., Fumeron F. Plasma Dihydroceramides are diabetes susceptibility biomarker candidates in mice and humans. Cell Rep. 2017;18:2269–2279.
    1. Braicu E.I., Darb-Esfahani S., Schmitt W.D., Koistinen K.M., Heiskanen L., Pöhö P. High-grade ovarian serous carcinoma patients exhibit profound alterations in lipid metabolism. Oncotarget. 2017;8:102912–102922.
    1. Chen L., Chen H., Li Y., Li L., Qiu Y., Ren J. Endocannabinoid and ceramide levels are altered in patients with colorectal cancer. Oncol Rep. 2015;34:447–454.
    1. Knapp P., Bodnar L., Błachnio-Zabielska A., Świderska M., Chabowski A. Plasma and ovarian tissue sphingolipids profiling in patients with advanced ovarian cancer. Gynecol Oncol. 2017;147:139–144.
    1. Kozar N., Kruusmaa K., Bitenc M., Argamasilla R., Adsuar A., Goswami N. Metabolomic profiling suggests long chain ceramides and sphingomyelins as a possible diagnostic biomarker of epithelial ovarian cancer. Clin Chim Acta. 2018;481:108–114.
    1. Separovic D., Shields A.F., Philip P.A., Bielawski J., Bielawska A., Pierce J.S. Altered levels of serum ceramide, sphingosine and sphingomyelin are associated with colorectal Cancer: a retrospective pilot study. Anticancer Res. 2017;37:1213–1218.
    1. Radner F.P.W., Marrakchi S., Kirchmeier P., Kim G.-J., Ribierre F., Kamoun B. Mutations in CERS3 cause autosomal recessive congenital ichthyosis in humans. PLoS Genet. 2013;9 e1003536.
    1. Kosinska M.K., Liebisch G., Lochnit G., Wilhelm J., Klein H., Kaesser U. Sphingolipids in human synovial fluid - a Lipidomic study. Plos One. 2014;9 e91769.
    1. Kosinska M.K., Mastbergen S.C., Liebisch G., Wilhelm J., Dettmeyer R.B., Ishaque B. Comparative lipidomic analysis of synovial fluid in human and canine osteoarthritis. Osteoarthr Cartil. 2016;24:1470–1478.
    1. Jiang X., Paultre F., Pearson T.A., Reed R.G., Francis C.K., Lin M. Plasma sphingomyelin level as a risk factor for coronary artery disease. Arterioscler Thromb Vasc Biol. 2000;20:2614–2618.
    1. Laaksonen R., Ekroos K., Sysi-Aho M., Hilvo M., Vihervaara T., Kauhanen D. Plasma ceramides predict cardiovascular death in patients with stable coronary artery disease and acute coronary syndromes beyond LDL-cholesterol. Eur Heart J. 2016;37:1967–1976.
    1. Meeusen Jeffrey W., Donato Leslie J., Bryant Sandra C., Baudhuin Linnea M., Berger Peter B., Jaffe Allan S. Plasma Ceramides. Arterioscler Thromb Vasc Biol. 2018;38:1933–1939.
    1. Yu J., Pan W., Shi R., Yang T., Li Y., Yu G. Ceramide is upregulated and associated with mortality in patients with chronic heart failure. Can J Cardiol. 2015;31:357–363.
    1. Abbott S.K., Li H., Muñoz S.S., Knoch B., Batterham M., Murphy K.E. Altered ceramide acyl chain length and ceramide synthase gene expression in Parkinson's disease. Mov Disord. 2014;29:518–526.
    1. Guedes L.C., Chan R.B., Gomes M.A., Conceição V.A., Machado R.B., Soares T. Serum lipid alterations in GBA-associated Parkinson's disease. Parkinsonism Relat Disord. 2017;44:58–65.
    1. Mielke M.M., Maetzler W., Haughey N.J., Bandaru V.V.R., Savica R., Deuschle C. Plasma ceramide and glucosylceramide metabolism is altered in sporadic Parkinson's disease and associated with cognitive impairment: a pilot study. PLoS One. 2013;8
    1. Xing Y., Tang Y., Zhao L., Wang Q., Qin W., Ji X. Associations between plasma ceramides and cognitive and neuropsychiatric manifestations in Parkinson's disease dementia. J Neurol Sci. 2016;370:82–87.
    1. Filippov V., Song M.A., Zhang K., Vinters H.V., Tung S., Kirsch W.M. Increased ceramide in brains with Alzheimer's and other neurodegenerative diseases. J Alzheimers Dis JAD. 2012;29:537–547.
    1. Kim M., Nevado-Holgado A., Whiley L., Snowden S.G., Soininen H., Kloszewska I. Association between plasma ceramides and phosphatidylcholines and hippocampal brain volume in late onset Alzheimer's disease. J Alzheimers Dis. 2017;60:809–817.
    1. Mielke M.M., Haughey N.J., Bandaru V.V.R., Schech S., Carrick R., Carlson M.C. Plasma ceramides are altered in mild cognitive impairment and predict cognitive decline and hippocampal volume loss. Alzheimers Dement. 2010;6:378–385.
    1. Mielke M.M., Haughey N.J., Bandaru V.V.R., Weinberg D.D., Darby E., Zaidi N. Plasma sphingomyelins are associated with cognitive progression in Alzheimer's disease. J Alzheimers Dis JAD. 2011;27:259–269.
    1. Mielke M.M., Bandaru V.V.R., Haughey N.J., Xia J., Fried L.P., Yasar S. Serum ceramides increase the risk of Alzheimer disease. Neurology. 2012;79:633.
    1. Hannun Y.A., Obeid L.M. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol. 2008;9:139–150.
    1. Hannun Y.A., Obeid L.M. Many ceramides. J Biol Chem. 2011;286:27855–27862.
    1. Chavez J.A., Summers S.A. Characterizing the effects of saturated fatty acids on insulin signaling and ceramide and diacylglycerol accumulation in 3T3-L1 adipocytes and C2C12 myotubes. Arch Biochem Biophys. 2003;419:101–109.
    1. O'Brien J.S., Sampson E.L. Lipid composition of the normal human brain: gray matter, white matter, and myelin. J Lipid Res. 1965;6:537–544.
    1. O'Brien J.S., Sampson E.L. Fatty acid and fatty aldehyde composition of the major brain lipids in normal human gray matter, white matter, and myelin. J Lipid Res. 1965;6:545–551.
    1. Jana A., Pahan K. Sphingolipids in multiple sclerosis. Neuromolecular Med. 2010;12:351–361.
    1. Kihara Y., Matsushita T., Kita Y., Uematsu S., Akira S., Kira J. Targeted lipidomics reveals mPGES-1-PGE2 as a therapeutic target for multiple sclerosis. Proc Natl Acad Sci. 2009;106:21807–21812.
    1. Jana A., Hogan E.L., Pahan K. Ceramide and neurodegeneration: susceptibility of neurons and oligodendrocytes to cell damage and death. J Neurol Sci. 2009;278:5–15.
    1. Mencarelli C., Martinez–Martinez P. Ceramide function in the brain: when a slight tilt is enough. Cell Mol Life Sci. 2013;70:181–203.
    1. Bieberich E. Ceramide signaling in cancer and stem cells. Future Lipidol. 2008;3:273–300.
    1. Kolesnick R. The therapeutic potential of modulating the ceramide/sphingomyelin pathway. J Clin Invest. 2002;110:3–8.
    1. Samanta S., Stiban J., Maugel T.K., Colombini M. Visualization of ceramide channels by transmission Electron microscopy. Biochim Biophys Acta. 2011;1808:1196–1201.
    1. Stiban J., Caputo L., Colombini M. Ceramide synthesis in the endoplasmic reticulum can permeabilize mitochondria to proapoptotic proteins. J Lipid Res. 2008;49:625–634.
    1. Baufeld C., O'Loughlin E., Calcagno N., Madore C., Butovsky O. Differential contribution of microglia and monocytes in neurodegenerative diseases. J Neural Transm. 2018;125:809–826.
    1. Lund H., Pieber M., Harris R.A. Lessons learned about neurodegeneration from microglia and monocyte depletion studies. Front Aging Neurosci. 2017;9
    1. Rawji K.S., Mishra M.K., Michaels N.J., Rivest S., Stys P.K., Yong V.W. Immunosenescence of microglia and macrophages: impact on the ageing central nervous system. Brain. 2016;139:653–661.
    1. Bitsch A., Schuchardt J., Bunkowski S., Kuhlmann T., Bruck W. Acute axonal injury in multiple sclerosis. Correlation with demyelination and inflammation. Brain J Neurol. 2000;123:1174–1183. Pt 6.
    1. Mishra M.K., Yong V.W. Myeloid cells — targets of medication in multiple sclerosis. Nat Rev Neurol. 2016;12:539–551.
    1. Nikić I., Merkler D., Sorbara C., Brinkoetter M., Kreutzfeldt M., Bareyre F.M. A reversible form of axon damage in experimental autoimmune encephalomyelitis and multiple sclerosis. Nat Med. 2011;17:495–499.
    1. Trapp B.D., Peterson J., Ransohoff R.M., Rudick R., Mörk S., Bö L. Axonal transection in the lesions of multiple sclerosis. N Engl J Med. 1998;338:278–285.
    1. Moreno M.A., Burns T., Yao P., Miers L., Pleasure D., Soulika A.M. Therapeutic depletion of monocyte-derived cells protects from long-term axonal loss in experimental autoimmune encephalomyelitis. J Neuroimmunol. 2016;290:36–46.

Source: PubMed

3
Iratkozz fel