Therapeutic efficacy of dimethyl fumarate in relapsing-remitting multiple sclerosis associates with ROS pathway in monocytes

Karl E Carlström, Ewoud Ewing, Mathias Granqvist, Alexandra Gyllenberg, Shahin Aeinehband, Sara Lind Enoksson, Antonio Checa, Tejaswi V S Badam, Jesse Huang, David Gomez-Cabrero, Mika Gustafsson, Faiez Al Nimer, Craig E Wheelock, Ingrid Kockum, Tomas Olsson, Maja Jagodic, Fredrik Piehl, Karl E Carlström, Ewoud Ewing, Mathias Granqvist, Alexandra Gyllenberg, Shahin Aeinehband, Sara Lind Enoksson, Antonio Checa, Tejaswi V S Badam, Jesse Huang, David Gomez-Cabrero, Mika Gustafsson, Faiez Al Nimer, Craig E Wheelock, Ingrid Kockum, Tomas Olsson, Maja Jagodic, Fredrik Piehl

Abstract

Dimethyl fumarate (DMF) is a first-line-treatment for relapsing-remitting multiple sclerosis (RRMS). The redox master regulator Nrf2, essential for redox balance, is a target of DMF, but its precise therapeutic mechanisms of action remain elusive. Here we show impact of DMF on circulating monocytes and T cells in a prospective longitudinal RRMS patient cohort. DMF increases the level of oxidized isoprostanes in peripheral blood. Other observed changes, including methylome and transcriptome profiles, occur in monocytes prior to T cells. Importantly, monocyte counts and monocytic ROS increase following DMF and distinguish patients with beneficial treatment-response from non-responders. A single nucleotide polymorphism in the ROS-generating NOX3 gene is associated with beneficial DMF treatment-response. Our data implicate monocyte-derived oxidative processes in autoimmune diseases and their treatment, and identify NOX3 genetic variant, monocyte counts and redox state as parameters potentially useful to inform clinical decisions on DMF therapy of RRMS.

Conflict of interest statement

F.P. has received research grants from Biogen, Novartis, and Genzyme, and fees for serving as Chair of DMC in clinical trials with Parexel. T.O. has received unrestricted MS research grants, and/or lecture advisory board honoraria from Biogen, Novartis, Sanofi and Roche. The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1
DMF induce increased isoprostane oxidation and transcription in response to oxidative stress. a Scheme of the study design. Peripheral blood was sampled at the clinic of patients fulfilling the criteria for RRMS and prescription of DMF. b Definition of RRMS patients as responders or non-responders to DMF therapy. c Experimental overlap between different assessments in the study d 8.12-iso-iPF2α-VI-isoprostane in plasma from paired patients sampled at three months (n = 9) and six months (n = 26) after DMF start compared to baseline (n = 26). e, f GSEA on mRNA from CD14+ sorted monocytes at baseline (n = 3) and six months (n = 3) shows an enrichment of upregulated genes involved in GO_REGULATION_OF_RESPONSE_TO_OXIDATIVE_STRESS at six months. ES, P-value and FDR were calculated by GSEA with weighted enrichment statistics and ratio of classes for the metric as input parameters. g Pathway analysis demonstrates oxidative stress-related canonical pathways in response to DMF (n = 3). Differentially expressed genes (P < 0.01 with average RMA > 4, one-way ANOVA) were used in IPA and significant pathways were determined with the right-tailed Fisher’s exact test (P < 0.05). The ratio indicates pathway’s activation status upon DMF treatment (g). Analysis in (d) was done by one-way ANOVA comparing both time points to baseline and graph shows mean and S.D. *P < 0.05, **P < 0.01
Fig. 2
Fig. 2
Monocyte numbers and ROS generation separate DMF responders from non-responders. a Representative plots for gating strategy of monocyte subsets stating mean ± S.E.M. at three months. Histogram shows representative DHR-123 MFI ± S.E.M. at 3 months. bd Number of monocyte subsets in DMF responders (n = 10) and non-responders (n = 10) at baseline and at three months. e Violin with box-and-whisker plots indicating values outside the 5–95 percentile are indicated as dots of monocytes and (f) lymphocytes from healthy controls (n = 28), DMF responders (n = 171) and DMF non-responders (n = 26) over time. g Spontaneous generation of ROS in healthy controls (n = 10) and DMF untreated (n = 18) and treated (n = 18) patients. h ROS generation in ex vivo-stimulated monocytes from DMF responders (n = 10) and non-responders (n = 7) measured with DHR-123. i Correlation between Δlymphocyte number and 8.12-iso-iPF2α-VI-isoprostanes at 6 months determined with Spearman’s correlation (n = 17). Graph (bd, g, h) shows mean and S.D. Analysis in (f) was performed with ANOVA for linear trend over time and (e) to test mean comparison between responders and non-responders at every timepoint. Remaining analysis between paired patients were performed with paired t test whereas analysis between healthy controls and patients or between patient groups were performed with Student’s two-tailed t test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001
Fig. 3
Fig. 3
Monocytes undergo DNA methylation changes at three months. a DNA methylation was measured using Illumina EPIC arrays in CD14+ monocytes sorted from peripheral blood of RRMS patients before (baseline; BL, n = 14) treatment with DMF as well as three and six months following the treatment (M3 n = 12 and M6 n = 9). Volcano plots illustrating differences in DNA methylation between different time points following DMF treatment. Hyper- and hypo-methylated CpGs with min 5% methylation change and P < 0.001 (Linear model testing) are indicated in red and blue, respectively. b Heat map of 1614 most significant differentially methylated CpG sites between the time points (the scale represents z-score). c Clusters of canonical pathways, derived using Ingenuity Pathway Analysis, suggest functions that are affected in monocytes following DMF treatment. The significance of pathways was determined with the right-tailed Fisher’s exact test (P < 0.05) and the clustering was performed on the relative risk for the overlap of molecules between the pathways using k-means. The horizontal top bar indicates the degree of overlap with selected ROS genes. Summary of the key functions and their constituent genes that displayed changes in DNA methylation upon DMF treatment are given to the right
Fig. 4
Fig. 4
Genetic association with SNPs in NOX3 to ROS generation and response to DMF treatment. a The G allele of rs6919626 (red line) was suggestively associated with reduced ROS generation in monocytes after ex vivo stimulation with E. coli (P = 0.057, β = −0.28) and significantly associated with lack of response to DMF treatment (P = 0.036, OR = 1.57). The Linkage Disequilibrium (LDplot) of the markers in NOX3 was generated with HaploView4.2 in the Swedish population. Darker gray indicates higher r2 between markers. b Methylation in four CpGs in the NOX3 promotor region over time and between responders (A allele) and non-responders (G allele) (Baseline: n = 6 + 6, three months: n = 5 + 6, six months: n = 6 + 1). c Schematic illustration indicating methylated or unmethylated CpGs in the NOX3 promotor region. dNOX3 transcription in responders and non-responders at six months following DMF (n = 5 + 4). Analysis performed using Welch’s two-tailed t test. Graph (b, d) shows box-and-whisker plots indicating values outside the 5–95 percentile are indicated as dots
Fig. 5
Fig. 5
DNA methylation changes are delayed in T cells and occur after six months. a DNA methylation was measured using Illumina EPIC arrays in CD4+ T cells sorted from peripheral blood of RRMS patients before (baseline; BL, n = 17) treatment with DMF as well as three and six months following the treatment M3 and M6 n = 12). Volcano plots illustrating differences in DNA methylation between different time points following DMF treatment. Hyper- and hypo-methylated CpGs with min 5% methylation change and P < 0.001 (Linear model testing) are indicated in red and blue, respectively. b Heat map of 1614 most significant differentially methylated CpG sites between the time points (the scale represents z-score). c Clusters of canonical pathways, derived using Ingenuity Pathway Analysis suggest functions that are affected in monocytes following six months of DMF treatment. The heat-map depicts cluster 1–3. The significance of pathways was determined with the right-tailed Fisher’s exact test (P < 0.05) and the clustering was performed on the relative risk for the overlap of molecules between the pathways using Mclust. The horizontal top bar indicates the degree of overlap with selected ROS genes. Summary of the key functions and their constituent genes that displayed changes in DNA methylation upon DMF treatment are given to the right
Fig. 6
Fig. 6
DMF responders show altered levels of CD4+ T cells subsets compared to non-responders. a, b Percentage and absolute number of naïve cells of CD4+ T cells was analyzed in DMF responders (n = 8) and non-responders (n = 4) at baseline and after six months. cf Central memory T cells (TCM) was defined as CD45RA−CCR7+ and effector memory T cells (TEM) as CD45RA−CCR7+. Graphs show means and S.D. and all analysis between paired patients are performed with paired t test. g, h Normalized protein expression (NPX) in plasma of IL17A, IL17C, CCL28, CDCP1, SLAM, EN-RAGE, IL12B, IL18R CXCL9 CCL4 and TWEAK were analyzed in responders (n = 29) and non-responders (n = 9) at baseline and 6 months after DMF intervention. *P < 0.05, **P < 0.01, ***P < 0.001. Asterisk in (d) indicate P value adjusted for multiple testing. **adjP < 0.01, ***adjP < 0.0001

References

    1. Suzuki T, Motohashi H, Yamamoto M. Toward clinical application of the Keap1-Nrf2 pathway. Trends Pharmacol. Sci. 2013;34:340–346. doi: 10.1016/j.tips.2013.04.005.
    1. Mossberg N, et al. Oxygen radical production in leukocytes and disease severity in multiple sclerosis. J. Neuroimmunol. 2009;213:131–134. doi: 10.1016/j.jneuroim.2009.05.013.
    1. Mix E, et al. Lymphocyte subpopulations, oxidative burst and apoptosis in peripheral blood cells of patients with multiple sclerosis-effect of interferon-beta. Autoimmunity. 2003;36:291–305. doi: 10.1080/0891693031000152697.
    1. Linker RA, et al. Fumaric acid esters exert neuroprotective effects in neuroinflammation via activation of the Nrf2 antioxidant pathway. Brain. 2011;134:678–692. doi: 10.1093/brain/awq386.
    1. Gopal S, et al. Evidence of activation of the Nrf2 pathway in multiple sclerosis patients treated with delayed-release dimethyl fumarate in the Phase 3 DEFINE and CONFIRM studies. Mult. Scler. 2017;23:1875–1883. doi: 10.1177/1352458517690617.
    1. Johansson K, et al. Cross talk in HEK293 cells between Nrf2, HIF, and NF-κB activities upon challenges with redox therapeutics characterized with single-cell resolution. Antioxid. Redox Signal. 2017;26:229–246. doi: 10.1089/ars.2015.6419.
    1. Huang H, Taraboletti A, Shriver LP. Dimethyl fumarate modulates antioxidant and lipid metabolism in oligodendrocytes. Redox Biol. 2015;5:169–175. doi: 10.1016/j.redox.2015.04.011.
    1. Scannevin RH, et al. Fumarates promote cytoprotection of central nervous system cells against oxidative stress via the nuclear factor (erythroid-derived 2)-like 2 pathway. J. Pharmacol. Exp. Ther. 2012;341:274–284. doi: 10.1124/jpet.111.190132.
    1. Borg G, et al. Auranofin improves outcome in early rheumatoid arthritis. Results from a 2-year, double blind placebo controlled study. J. Rheuma. 1988;15:1747–1754.
    1. Huck F, de Médicis R, Lussier A, Dupuis G, Federlin P. Reducing property of some slow acting antirheumatic drugs. J. Rheumatol. 1984;11:605–609.
    1. Madeira JM, Gibson DL, Kean WF, Klegeris A. The biological activity of auranofin: implications for novel treatment of diseases. Inflammopharmacology. 2012;20:297–306. doi: 10.1007/s10787-012-0149-1.
    1. Moharregh-Khiabani D, et al. Effects of fumaric acids on cuprizone induced central nervous system de- and remyelination in the mouse. PLoS ONE. 2010;5:e11769. doi: 10.1371/journal.pone.0011769.
    1. Palmer AM. Multiple sclerosis and the blood-central nervous system barrier. Cardiovasc. Psychiatry Neurol. 2013;2013:530356–10. doi: 10.1155/2013/530356.
    1. Gross CC, et al. Dimethyl fumarate treatment alters circulating T helper cell subsets in multiple sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2016;3:e183. doi: 10.1212/NXI.0000000000000183.
    1. Spencer CM, Crabtree-Hartman EC, Lehmann-Horn K, Cree BAC, Zamvil SS. Reduction of CD8( + ) T lymphocytes in multiple sclerosis patients treated with dimethyl fumarate. Neurol. Neuroimmunol. Neuroinflamm. 2015;2:e76. doi: 10.1212/NXI.0000000000000076.
    1. Khatri BO, et al. The effect of dimethyl fumarate (Tecfidera™) on lymphocyte counts: a potential contributor to progressive multifocal leukoencephalopathy risk. Mult. Scler. Relat. Disord. 2015;4:377–379. doi: 10.1016/j.msard.2015.05.003.
    1. Hammer A, et al. The NRF2 pathway as potential biomarker for dimethyl fumarate treatment in multiple sclerosis. Ann. Clin. Transl. Neurol. 2018;5:668–676. doi: 10.1002/acn3.553.
    1. Wu Q, et al. Dimethyl fumarate selectively reduces memory T cells and shifts the balance between Th1/Th17 and Th2 in multiple sclerosis patients. J. Immunol. 2017;198:3069–3080. doi: 10.4049/jimmunol.1601532.
    1. Ghoreschi K, et al. Fumarates improve psoriasis and multiple sclerosis by inducing type II dendritic cells. J. Exp. Med. 2011;208:2291–2303. doi: 10.1084/jem.20100977.
    1. Gold R, et al. Placebo-controlled phase 3 study of oral BG-12 for relapsing multiple sclerosis. N. Engl. J. Med. 2012;367:1098–1107. doi: 10.1056/NEJMoa1114287.
    1. Fox RJ, et al. Placebo-controlled phase 3 study of oral BG-12 or glatiramer in multiple sclerosis. N. Engl. J. Med. 2012;367:1087–1097. doi: 10.1056/NEJMoa1206328.
    1. Hultqvist M, et al. Enhanced autoimmunity, arthritis, and encephalomyelitis in mice with a reduced oxidative burst due to a mutation in the Ncf1 gene. Proc. Natl Acad. Sci. USA. 2004;101:12646–12651. doi: 10.1073/pnas.0403831101.
    1. Belikov AV, Schraven B, Simeoni L. T cells and reactive oxygen species. J. Biomed. Sci. 2015;22:85. doi: 10.1186/s12929-015-0194-3.
    1. Holmdahl R, et al. Hydrogen peroxide as an immunological transmitter regulating autoreactive T cells. Antioxid. Redox Signal. 2013;18:1463–1474. doi: 10.1089/ars.2012.4734.
    1. Olofsson P, et al. Positional identification of Ncf1 as a gene that regulates arthritis severity in rats. Nat. Genet. 2003;33:25–32. doi: 10.1038/ng1058.
    1. Becanovic K, et al. Advanced intercross line mapping of Eae5 reveals Ncf-1 and CLDN4 as candidate genes for experimental autoimmune encephalomyelitis. J. Immunol. 2006;176:6055–6064. doi: 10.4049/jimmunol.176.10.6055.
    1. van der Veen RC, et al. Superoxide prevents nitric oxide-mediated suppression of helper T lymphocytes: decreased autoimmune encephalomyelitis in nicotinamide adenine dinucleotide phosphate oxidase knockout mice. J. Immunol. 2000;164:5177–5183. doi: 10.4049/jimmunol.164.10.5177.
    1. Kraaij MD, et al. Induction of regulatory T cells by macrophages is dependent on production of reactive oxygen species. Proc. Natl Acad. Sci. USA. 2010;107:17686–17691. doi: 10.1073/pnas.1012016107.
    1. Lee K, Won HY, Bae MA, Hong J-H, Hwang ES. Spontaneous and aging-dependent development of arthritis in NADPH oxidase 2 deficiency through altered differentiation of CD11b + and Th/Treg cells. Proc. Natl Acad. Sci. USA. 2011;108:9548–9553. doi: 10.1073/pnas.1012645108.
    1. Klemke M, et al. Oxidation of cofilin mediates T cell hyporesponsiveness under oxidative stress conditions. Immunity. 2008;29:404–413. doi: 10.1016/j.immuni.2008.06.016.
    1. Tse HM, et al. NADPH oxidase deficiency regulates Th lineage commitment and modulates autoimmunity. J. Immunol. 2010;185:5247–5258. doi: 10.4049/jimmunol.1001472.
    1. Kwon J, et al. The nonphagocytic NADPH oxidase Duox1 mediates a positive feedback loop during T cell receptor signaling. Sci. Signal. 2010;3:ra59–ra59. doi: 10.1126/scisignal.2000976.
    1. Cemerski S, Cantagrel A, Van Meerwijk JPM, Romagnoli P. Reactive oxygen species differentially affect T cell receptor-signaling pathways. J. Biol. Chem. 2002;277:19585–19593. doi: 10.1074/jbc.M111451200.
    1. Simeoni L, Bogeski I. Redox regulation of T-cell receptor signaling. Biol. Chem. 2015;396:555–568. doi: 10.1515/hsz-2014-0312.
    1. Schmielau J, Finn OJ. Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of t-cell function in advanced cancer patients. Cancer Res. 2001;61:4756–4760.
    1. George-Chandy A, et al. Th17 development and autoimmune arthritis in the absence of reactive oxygen species. Eur. J. Immunol. 2008;38:1118–1126. doi: 10.1002/eji.200737348.
    1. Fu G, et al. Suppression of Th17 cell differentiation by misshapen/NIK-related kinase MINK1. J. Exp. Med. 2017;214:1453–1469. doi: 10.1084/jem.20161120.
    1. Yi H, Guo C, Yu X, Zuo D, Wang X-Y. Mouse CD11b + Gr-1 + myeloid cells can promote Th17 cell differentiation and experimental autoimmune encephalomyelitis. J. Immunol. 2012;189:4295–4304. doi: 10.4049/jimmunol.1200086.
    1. Mougiakakos D, Johansson CC, Kiessling R. Naturally occurring regulatory T cells show reduced sensitivity toward oxidative stress-induced cell death. Blood. 2009;113:3542–3545. doi: 10.1182/blood-2008-09-181040.
    1. Bedard K, Krause K-H. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol. Rev. 2007;87:245–313. doi: 10.1152/physrev.00044.2005.
    1. Lee CF, Qiao M, Schröder K, Zhao Q, Asmis R. Nox4 is a novel inducible source of reactive oxygen species in monocytes and macrophages and mediates oxidized low density lipoprotein-induced macrophage death. Circ. Res. 2010;106:1489–1497. doi: 10.1161/CIRCRESAHA.109.215392.
    1. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009;9:162–174. doi: 10.1038/nri2506.
    1. Brown KL, et al. ROS-deficient monocytes have aberrant gene expression that correlates with inflammatory disorders of chronic granulomatous disease. Clin. Immunol. 2008;129:90–102. doi: 10.1016/j.clim.2008.06.005.
    1. Noubade R, et al. NRROS negatively regulates reactive oxygen species during host defence and autoimmunity. Nature. 2014;509:235–239. doi: 10.1038/nature13152.
    1. van Horssen J, et al. NAD(P)H:quinone oxidoreductase 1 expression in multiple sclerosis lesions. Free Radic. Biol. Med. 2006;41:311–317. doi: 10.1016/j.freeradbiomed.2006.04.013.
    1. Fischer MT, et al. NADPH oxidase expression in active multiple sclerosis lesions in relation to oxidative tissue damage and mitochondrial injury. Brain. 2012;135:886–899. doi: 10.1093/brain/aws012.
    1. Shu Y, et al. Epigenetic variability of CD4 + CD25 + Tregs contributes to the pathogenesis of autoimmune diseases. Clin. Rev. Allergy Immunol. 2017;52:260–272. doi: 10.1007/s12016-016-8590-3.
    1. Ray D, Yung R. Immune senescence, epigenetics and autoimmunity. Clin. Immunol. 2018;196:59–63. doi: 10.1016/j.clim.2018.04.002.
    1. Montuschi P, Barnes PJ, Roberts LJ. Isoprostanes: markers and mediators of oxidative stress. FASEB J. 2004;18:1791–1800. doi: 10.1096/fj.04-2330rev.
    1. Milne GL, Yin H, Hardy KD, Davies SS, Roberts LJ. Isoprostane generation and function. Chem. Rev. 2011;111:5973–5996. doi: 10.1021/cr200160h.
    1. Zerbino DR, Wilder SP, Johnson N, Juettemann T, Flicek PR. The ensembl regulatory build. Genome Biol. 2015;16:56. doi: 10.1186/s13059-015-0621-5.
    1. Kesarwani P, Murali AK, Al-Khami AA, Mehrotra S. Redox regulation of T-cell function: from molecular mechanisms to significance in human health and disease. Antioxid. Redox Signal. 2013;18:1497–1534. doi: 10.1089/ars.2011.4073.
    1. Ohl K, Tenbrock K, Kipp M. Oxidative stress in multiple sclerosis: central and peripheral mode of action. Exp. Neurol. 2016;277:58–67. doi: 10.1016/j.expneurol.2015.11.010.
    1. Piehl F. A changing treatment landscape for multiple sclerosis: challenges and opportunities. J. Intern. Med. 2014;275:364–381. doi: 10.1111/joim.12204.
    1. Zhao C, et al. The CD14( + /low)CD16( + ) monocyte subset is more susceptible to spontaneous and oxidant-induced apoptosis than the CD14( + )CD16(-) subset. Cell Death Dis. 2010;1:e95–e95. doi: 10.1038/cddis.2010.69.
    1. Chen H, et al. Hydroxycarboxylic acid receptor 2 mediates dimethyl fumarate’s protective effect in EAE. J. Clin. Invest. 2014;124:2188–2192. doi: 10.1172/JCI72151.
    1. Wang Z, Chang C, Lu Q. Epigenetics of CD4 + T cells in autoimmune diseases. Curr. Opin. Rheuma. 2017;29:361–368. doi: 10.1097/BOR.0000000000000393.
    1. He H, Hu Z, Xiao H, Zhou F, Yang B. The tale of histone modifications and its role in multiple sclerosis. Hum. Genom. 2018;12:31. doi: 10.1186/s40246-018-0163-5.
    1. Patsopoulos, N. et al. The Multiple Sclerosis Genomic Map: Role of peripheral immune cells and resident microglia in susceptibility. bioRxiv 14, 143–933 (2017).
    1. Polman CH, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann. Neurol. 2011;69:292–302. doi: 10.1002/ana.22366.
    1. Maecker HT, McCoy JP, Nussenblatt R. Standardizing immunophenotyping for the Human Immunology Project. Nat. Rev. Immunol. 2012;12:191–200. doi: 10.1038/nri3158.
    1. Kolmert J, et al. Lipid mediator quantification in isolated human and guinea pig airways: an expanded approach for respiratory research. Anal. Chem. 2018;90:10239–10248. doi: 10.1021/acs.analchem.8b01651.
    1. Medina S, et al. A ultra-pressure liquid chromatography/triple quadrupole tandem mass spectrometry method for the analysis of 13 eicosanoids in human urine and quantitative 24 hour values in healthy volunteers in a controlled constant diet. Rapid Commun. Mass Spectrom. 2012;26:1249–1257. doi: 10.1002/rcm.6224.
    1. Morris TJ, et al. Bioinformatics. 2014. ChAMP: 450k chip analysis methylation pipeline; pp. 428–430.
    1. Marabita F, et al. An evaluation of analysis pipelines for DNA methylation profiling using the Illumina HumanMethylation450 BeadChip platform. Epigenetics. 2013;8:333–346. doi: 10.4161/epi.24008.
    1. Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28:882–883. doi: 10.1093/bioinformatics/bts034.
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47–e47. doi: 10.1093/nar/gkv007.
    1. Houseman EA, et al. Reference-free deconvolution of DNA methylation data and mediation by cell composition effects. BMC Bioinforma. 2016;17:259. doi: 10.1186/s12859-016-1140-4.

Source: PubMed

3
Suscribir