메뉴 건너뛰기




Volumn 113, Issue 41, 2016, Pages E6145-E6152

Laquinimod arrests experimental autoimmune encephalomyelitis by activating the aryl hydrocarbon receptor

Author keywords

Aryl hydrocarbon receptor; EAE; Laquinimod

Indexed keywords

3 METHYLCHOLANTHRENE; AROMATIC HYDROCARBON RECEPTOR; CYTOCHROME P450 1A; CYTOCHROME P450 1A1; CYTOCHROME P450 1A2; CYTOCHROME P450 1B1; INTERLEUKIN 17; INTERLEUKIN 22; LAQUINIMOD; MESSENGER RNA; MYELIN OLIGODENDROCYTE GLYCOPROTEIN; OMEPRAZOLE; QUINOLONE DERIVATIVE; TRANSCRIPTOME;

EID: 84991503343     PISSN: 00278424     EISSN: 10916490     Source Type: Journal    
DOI: 10.1073/pnas.1607843113     Document Type: Article
Times cited : (105)

References (51)
  • 1
    • 84858217865 scopus 로고    scopus 로고
    • Placebo-controlled trial of oral laquinimod for multiple sclerosis
    • Comi G, et al.; ALLEGRO Study Group (2012) Placebo-controlled trial of oral laquinimod for multiple sclerosis. N Engl J Med 366(11):1000-1009.
    • (2012) N Engl J Med , vol.366 , Issue.11 , pp. 1000-1009
    • Comi, G.1
  • 2
    • 84906537932 scopus 로고    scopus 로고
    • Placebo-controlled trial of oral laquinimod in multiple sclerosis: MRI evidence of an effect on brain tissue damage
    • Filippi M, et al.; ALLEGRO Study Group (2014) Placebo-controlled trial of oral laquinimod in multiple sclerosis: MRI evidence of an effect on brain tissue damage. J Neurol Neurosurg Psychiatry 85(8):851-858.
    • (2014) J Neurol Neurosurg Psychiatry , vol.85 , Issue.8 , pp. 851-858
    • Filippi, M.1
  • 3
    • 84898896452 scopus 로고    scopus 로고
    • A randomized placebo-controlled phase III trial of oral laquinimod for multiple sclerosis
    • Vollmer TL, et al.; BRAVO Study Group (2014) A randomized placebo-controlled phase III trial of oral laquinimod for multiple sclerosis. J Neurol 261(4):773-783.
    • (2014) J Neurol , vol.261 , Issue.4 , pp. 773-783
    • Vollmer, T.L.1
  • 4
    • 77956649428 scopus 로고    scopus 로고
    • Laquinimod interferes with migratory capacity of T cells and reduces IL-17 levels, inflammatory demyelination and acute axonal damage in mice with experimental autoimmune encephalomyelitis
    • Wegner C, et al. (2010) Laquinimod interferes with migratory capacity of T cells and reduces IL-17 levels, inflammatory demyelination and acute axonal damage in mice with experimental autoimmune encephalomyelitis. J Neuroimmunol 227(1-2):133-143.
    • (2010) J Neuroimmunol , vol.227 , Issue.1-2 , pp. 133-143
    • Wegner, C.1
  • 5
    • 84865631399 scopus 로고    scopus 로고
    • Oral treatment with laquinimod augments regulatory T-cells and brain-derived neurotrophic factor expression and reduces injury in the CNS of mice with experimental autoimmune encephalomyelitis
    • Aharoni R, et al. (2012) Oral treatment with laquinimod augments regulatory T-cells and brain-derived neurotrophic factor expression and reduces injury in the CNS of mice with experimental autoimmune encephalomyelitis. J Neuroimmunol 251(1-2):14-24.
    • (2012) J Neuroimmunol , vol.251 , Issue.1-2 , pp. 14-24
    • Aharoni, R.1
  • 6
    • 84864117759 scopus 로고    scopus 로고
    • Kinetics of proinflammatory monocytes in a model of multiple sclerosis and its perturbation by laquinimod
    • Mishra MK, Wang J, Silva C, Mack M, Yong VW (2012) Kinetics of proinflammatory monocytes in a model of multiple sclerosis and its perturbation by laquinimod. Am J Pathol 181(2):642-651.
    • (2012) Am J Pathol , vol.181 , Issue.2 , pp. 642-651
    • Mishra, M.K.1    Wang, J.2    Silva, C.3    Mack, M.4    Yong, V.W.5
  • 7
    • 84859151160 scopus 로고    scopus 로고
    • Laquinimod, a quinoline-3-carboxamide, induces type II myeloid cells that modulate central nervous system autoimmunity
    • Schulze-Topphoff U, et al. (2012) Laquinimod, a quinoline-3-carboxamide, induces type II myeloid cells that modulate central nervous system autoimmunity. PLoS One 7(3):e33797.
    • (2012) PLoS One , vol.7 , Issue.3 , pp. e33797
    • Schulze-Topphoff, U.1
  • 8
    • 83555174399 scopus 로고    scopus 로고
    • Modulation of autoimmune demyelination by laquinimod via induction of brain-derived neurotrophic factor
    • Thone J, et al. (2012) Modulation of autoimmune demyelination by laquinimod via induction of brain-derived neurotrophic factor. Am J Pathol 180(1):267-274.
    • (2012) Am J Pathol , vol.180 , Issue.1 , pp. 267-274
    • Thone, J.1
  • 9
    • 84876042951 scopus 로고    scopus 로고
    • Modulation of dendritic cell properties by laquinimod as a mechanism for modulating multiple sclerosis
    • Jolivel V, et al. (2013) Modulation of dendritic cell properties by laquinimod as a mechanism for modulating multiple sclerosis. Brain 136(Pt 4):1048-1066.
    • (2013) Brain , vol.136 , pp. 1048-1066
    • Jolivel, V.1
  • 10
    • 84879758332 scopus 로고    scopus 로고
    • Laquinimod prevents inflammation-induced synaptic alterations occurring in experimental autoimmune encephalomyelitis
    • Ruffini F, et al. (2013) Laquinimod prevents inflammation-induced synaptic alterations occurring in experimental autoimmune encephalomyelitis. Mult Scler 19(8):1084-1094.
    • (2013) Mult Scler , vol.19 , Issue.8 , pp. 1084-1094
    • Ruffini, F.1
  • 11
    • 84893507025 scopus 로고    scopus 로고
    • Therapeutic laquinimod treatment decreases inflammation, initiates axon remyelination, and improves motor deficit in a mouse model of multiple sclerosis
    • Moore S, et al. (2013) Therapeutic laquinimod treatment decreases inflammation, initiates axon remyelination, and improves motor deficit in a mouse model of multiple sclerosis. Brain Behav 3(6):664-682.
    • (2013) Brain Behav , vol.3 , Issue.6 , pp. 664-682
    • Moore, S.1
  • 12
    • 84922986271 scopus 로고    scopus 로고
    • Laquinimod reduces neuroaxonal injury through inhibiting microglial activation
    • Mishra MK, et al. (2014) Laquinimod reduces neuroaxonal injury through inhibiting microglial activation. Ann Clin Transl Neurol 1(6):409-422.
    • (2014) Ann Clin Transl Neurol , vol.1 , Issue.6 , pp. 409-422
    • Mishra, M.K.1
  • 13
    • 0036240732 scopus 로고    scopus 로고
    • Suppression of experimental autoimmune neuritis by ABR-215062 is associated with altered Th1/Th2 balance and inhibited migration of inflammatory cells into the peripheral nerve tissue
    • Zou LP, et al. (2002) Suppression of experimental autoimmune neuritis by ABR-215062 is associated with altered Th1/Th2 balance and inhibited migration of inflammatory cells into the peripheral nerve tissue. Neuropharmacology 42(5):731-739.
    • (2002) Neuropharmacology , vol.42 , Issue.5 , pp. 731-739
    • Zou, L.P.1
  • 14
    • 84908666347 scopus 로고    scopus 로고
    • Laquinimod exerts strong clinical and immunomodulatory effects in Lewis rat experimental autoimmune neuritis
    • Pitarokoili K, et al. (2014) Laquinimod exerts strong clinical and immunomodulatory effects in Lewis rat experimental autoimmune neuritis. J Neuroimmunol 274(1-2):38-45.
    • (2014) J Neuroimmunol , vol.274 , Issue.1-2 , pp. 38-45
    • Pitarokoili, K.1
  • 15
    • 84896289833 scopus 로고    scopus 로고
    • Laquinimod delays and suppresses nephritis in lupus-prone mice and affects both myeloid and lymphoid immune cells
    • Lourenco EV, Wong M, Hahn BH, Palma-Diaz MF, Skaggs BJ (2014) Laquinimod delays and suppresses nephritis in lupus-prone mice and affects both myeloid and lymphoid immune cells. Arthritis Rheumatol 66(3):674-685.
    • (2014) Arthritis Rheumatol , vol.66 , Issue.3 , pp. 674-685
    • Lourenco, E.V.1    Wong, M.2    Hahn, B.H.3    Palma-Diaz, M.F.4    Skaggs, B.J.5
  • 16
    • 84948455050 scopus 로고    scopus 로고
    • Laquinimod ameliorates spontaneous colitis in interleukin-10-gene-deficient mice with improved barrier function
    • Sun J, et al. (2015) Laquinimod ameliorates spontaneous colitis in interleukin-10-gene-deficient mice with improved barrier function. Int Immunopharmacol 29(2):423-432.
    • (2015) Int Immunopharmacol , vol.29 , Issue.2 , pp. 423-432
    • Sun, J.1
  • 17
    • 84908655755 scopus 로고    scopus 로고
    • Laquinimod, an up-and-coming immunomodulatory agent for treatment of multiple sclerosis
    • Varrin-Doyer M, Zamvil SS, Schulze-Topphoff U (2014) Laquinimod, an up-and-coming immunomodulatory agent for treatment of multiple sclerosis. Exp Neurol 262(Pt A):66-71.
    • (2014) Exp Neurol , vol.262 , pp. 66-71
    • Varrin-Doyer, M.1    Zamvil, S.S.2    Schulze-Topphoff, U.3
  • 18
    • 34347327220 scopus 로고    scopus 로고
    • Induction of cyp1a1 is a nonspecific biomarker of aryl hydrocarbon receptor activation: Results of large scale screening of pharmaceuticals and toxicants in vivo and in vitro
    • Hu W, Sorrentino C, Denison MS, Kolaja K, Fielden MR (2007) Induction of cyp1a1 is a nonspecific biomarker of aryl hydrocarbon receptor activation: Results of large scale screening of pharmaceuticals and toxicants in vivo and in vitro. Mol Pharmacol 71(6):1475-1486.
    • (2007) Mol Pharmacol , vol.71 , Issue.6 , pp. 1475-1486
    • Hu, W.1    Sorrentino, C.2    Denison, M.S.3    Kolaja, K.4    Fielden, M.R.5
  • 19
    • 84899989767 scopus 로고    scopus 로고
    • Aryl hydrocarbon receptor repressor and TiPARP (ARTD14) use similar, but also distinct mechanisms to repress aryl hydrocarbon receptor signaling
    • MacPherson L, et al. (2014) Aryl hydrocarbon receptor repressor and TiPARP (ARTD14) use similar, but also distinct mechanisms to repress aryl hydrocarbon receptor signaling. Int J Mol Sci 15(5):7939-7957.
    • (2014) Int J Mol Sci , vol.15 , Issue.5 , pp. 7939-7957
    • MacPherson, L.1
  • 20
    • 79851516167 scopus 로고    scopus 로고
    • Hepatic transcriptomic responses to TCDD in dioxin-sensitive and dioxin-resistant rats during the onset of toxicity
    • Boutros PC, et al. (2011) Hepatic transcriptomic responses to TCDD in dioxin-sensitive and dioxin-resistant rats during the onset of toxicity. Toxicol Appl Pharmacol 251(2):119-129.
    • (2011) Toxicol Appl Pharmacol , vol.251 , Issue.2 , pp. 119-129
    • Boutros, P.C.1
  • 21
    • 84918787819 scopus 로고    scopus 로고
    • Immunological relevance of the coevolution of IDO1 and AHR
    • Jaronen M, Quintana FJ (2014) Immunological relevance of the coevolution of IDO1 and AHR. Front Immunol 5:5-1.
    • (2014) Front Immunol , vol.5 , pp. 5-11
    • Jaronen, M.1    Quintana, F.J.2
  • 22
    • 84919427790 scopus 로고    scopus 로고
    • Aryl hydrocarbon receptor and kynurenine: Recent advances in autoimmune disease research
    • Nguyen NT, et al. (2014) Aryl hydrocarbon receptor and kynurenine: Recent advances in autoimmune disease research. Front Immunol 5:5-1.
    • (2014) Front Immunol , vol.5 , pp. 5-11
    • Nguyen, N.T.1
  • 23
    • 84869849711 scopus 로고    scopus 로고
    • Xenobiotics and loss of cell adhesion drive distinct transcriptional outcomes by aryl hydrocarbon receptor signaling
    • Hao N, et al. (2012) Xenobiotics and loss of cell adhesion drive distinct transcriptional outcomes by aryl hydrocarbon receptor signaling. Mol Pharmacol 82(6):1082-1093.
    • (2012) Mol Pharmacol , vol.82 , Issue.6 , pp. 1082-1093
    • Hao, N.1
  • 24
    • 78650547540 scopus 로고    scopus 로고
    • An endogenous aryl hydrocarbon receptor ligand acts on dendritic cells and T cells to suppress experimental autoimmune encephalomyelitis
    • Quintana FJ, et al. (2010) An endogenous aryl hydrocarbon receptor ligand acts on dendritic cells and T cells to suppress experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA 107(48):20768-20773.
    • (2010) Proc Natl Acad Sci USA , vol.107 , Issue.48 , pp. 20768-20773
    • Quintana, F.J.1
  • 25
    • 42649095378 scopus 로고    scopus 로고
    • Control of T (reg) and T (H) 17 cell differentiation by the aryl hydrocarbon receptor
    • Quintana FJ, et al. (2008) Control of T (reg) and T (H) 17 cell differentiation by the aryl hydrocarbon receptor. Nature 453(7191):65-71.
    • (2008) Nature , vol.453 , Issue.7191 , pp. 65-71
    • Quintana, F.J.1
  • 26
    • 47749107262 scopus 로고    scopus 로고
    • Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells
    • Kimura A, Naka T, Nohara K, Fujii-Kuriyama Y, Kishimoto T (2008) Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells. Proc Natl Acad Sci USA 105(28):9721-9726.
    • (2008) Proc Natl Acad Sci USA , vol.105 , Issue.28 , pp. 9721-9726
    • Kimura, A.1    Naka, T.2    Nohara, K.3    Fujii-Kuriyama, Y.4    Kishimoto, T.5
  • 27
    • 84893938271 scopus 로고    scopus 로고
    • Differential influences of the aryl hydrocarbon receptor on Th17 mediated responses in vitro and in vivo
    • Duarte JH, Di Meglio P, Hirota K, Ahlfors H, Stockinger B (2013) Differential influences of the aryl hydrocarbon receptor on Th17 mediated responses in vitro and in vivo. PLoS One 8(11):e79819.
    • (2013) PLoS One , vol.8 , Issue.11 , pp. e79819
    • Duarte, J.H.1    Di Meglio, P.2    Hirota, K.3    Ahlfors, H.4    Stockinger, B.5
  • 28
    • 84928497841 scopus 로고    scopus 로고
    • The aryl hydrocarbon receptor: Differential contribution to T helper 17 and T cytotoxic 17 cell development
    • Hayes MD, Ovcinnikovs V, Smith AG, Kimber I, Dearman RJ (2014) The aryl hydrocarbon receptor: Differential contribution to T helper 17 and T cytotoxic 17 cell development. PLoS One 9(9):e106955.
    • (2014) PLoS One , vol.9 , Issue.9 , pp. e106955
    • Hayes, M.D.1    Ovcinnikovs, V.2    Smith, A.G.3    Kimber, I.4    Dearman, R.J.5
  • 29
    • 84886096349 scopus 로고    scopus 로고
    • Regulation of central nervous system autoimmunity by the aryl hydrocarbon receptor
    • Quintana FJ (2013) Regulation of central nervous system autoimmunity by the aryl hydrocarbon receptor. Semin Immunopathol 35(6):627-635.
    • (2013) Semin Immunopathol , vol.35 , Issue.6 , pp. 627-635
    • Quintana, F.J.1
  • 30
    • 84863493136 scopus 로고    scopus 로고
    • Aryl hydrocarbon receptor controls regulatory CD4+ T cell function
    • Pot C (2012) Aryl hydrocarbon receptor controls regulatory CD4+ T cell function. Swiss Med Wkly 142:w13592.
    • (2012) Swiss Med Wkly , vol.142 , pp. w13592
    • Pot, C.1
  • 31
    • 77955903212 scopus 로고    scopus 로고
    • The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27
    • Apetoh L, et al. (2010) The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat Immunol 11(9):854-861.
    • (2010) Nat Immunol , vol.11 , Issue.9 , pp. 854-861
    • Apetoh, L.1
  • 32
    • 77955859470 scopus 로고    scopus 로고
    • Activation of the aryl hydrocarbon receptor induces human type 1 regulatory T cell-like and Foxp3 (+) regulatory T cells
    • Gandhi R, et al. (2010) Activation of the aryl hydrocarbon receptor induces human type 1 regulatory T cell-like and Foxp3(+) regulatory T cells. Nat Immunol 11(9):846-853.
    • (2010) Nat Immunol , vol.11 , Issue.9 , pp. 846-853
    • Gandhi, R.1
  • 33
    • 80051968211 scopus 로고    scopus 로고
    • In vivo induction of Tr1 cells via mucosal dendritic cells and AHR signaling
    • Wu HY, et al. (2011) In vivo induction of Tr1 cells via mucosal dendritic cells and AHR signaling. PLoS One 6(8):e23618.
    • (2011) PLoS One , vol.6 , Issue.8 , pp. e23618
    • Wu, H.Y.1
  • 34
    • 84930576092 scopus 로고    scopus 로고
    • Metabolic control of type 1 regulatory T cell differentiation by AHR and HIF1-α
    • Mascanfroni ID, et al. (2015) Metabolic control of type 1 regulatory T cell differentiation by AHR and HIF1-α. Nat Med 21(6):638-646.
    • (2015) Nat Med , vol.21 , Issue.6 , pp. 638-646
    • Mascanfroni, I.D.1
  • 35
    • 84155196846 scopus 로고    scopus 로고
    • Exactly the same but different: Promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor
    • Denison MS, Soshilov AA, He G, DeGroot DE, Zhao B (2011) Exactly the same but different: Promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol Sci 124(1):1-22.
    • (2011) Toxicol Sci , vol.124 , Issue.1 , pp. 1-22
    • Denison, M.S.1    Soshilov, A.A.2    He, G.3    DeGroot, D.E.4    Zhao, B.5
  • 36
    • 84865863065 scopus 로고    scopus 로고
    • Reduced astrocytic NF-κB activation by laquinimod protects from cuprizone-induced demyelination
    • Bruck W, et al. (2012) Reduced astrocytic NF-κB activation by laquinimod protects from cuprizone-induced demyelination. Acta Neuropathol 124(3):411-424.
    • (2012) Acta Neuropathol , vol.124 , Issue.3 , pp. 411-424
    • Bruck, W.1
  • 37
    • 0036765531 scopus 로고    scopus 로고
    • 2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin suppresses tumor necrosis factor-alpha and anti-CD40-induced activation of NF-kappaB/Rel in dendritic cells: P50 homodimer activation is not affected
    • Ruby CE, Leid M, Kerkvliet NI (2002) 2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin suppresses tumor necrosis factor-alpha and anti-CD40-induced activation of NF-kappaB/Rel in dendritic cells: p50 homodimer activation is not affected. Mol Pharmacol 62(3):722-728.
    • (2002) Mol Pharmacol , vol.62 , Issue.3 , pp. 722-728
    • Ruby, C.E.1    Leid, M.2    Kerkvliet, N.I.3
  • 38
    • 84892642621 scopus 로고    scopus 로고
    • Cross-talk between aryl hydrocarbon receptor and the inflammatory response: A role for nuclear factor-κB
    • Vogel CF, et al. (2014) Cross-talk between aryl hydrocarbon receptor and the inflammatory response: A role for nuclear factor-κB. J Biol Chem 289(3):1866-1875.
    • (2014) J Biol Chem , vol.289 , Issue.3 , pp. 1866-1875
    • Vogel, C.F.1
  • 39
    • 84966658995 scopus 로고    scopus 로고
    • Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor
    • Rothhammer V, et al. (2016) Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat Med 22(6):586-597.
    • (2016) Nat Med , vol.22 , Issue.6 , pp. 586-597
    • Rothhammer, V.1
  • 40
    • 84922377460 scopus 로고    scopus 로고
    • The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology
    • Esser C, Rannug A (2015) The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacol Rev 67(2):259-279.
    • (2015) Pharmacol Rev , vol.67 , Issue.2 , pp. 259-279
    • Esser, C.1    Rannug, A.2
  • 41
    • 84878454583 scopus 로고    scopus 로고
    • The roles of aryl hydrocarbon receptor in immune responses
    • Nguyen NT, Hanieh H, Nakahama T, Kishimoto T (2013) The roles of aryl hydrocarbon receptor in immune responses. Int Immunol 25(6):335-343.
    • (2013) Int Immunol , vol.25 , Issue.6 , pp. 335-343
    • Nguyen, N.T.1    Hanieh, H.2    Nakahama, T.3    Kishimoto, T.4
  • 42
    • 84888169376 scopus 로고    scopus 로고
    • Aryl hydrocarbon receptor control of adaptive immunity
    • Quintana FJ, Sherr DH (2013) Aryl hydrocarbon receptor control of adaptive immunity. Pharmacol Rev 65(4):1148-1161.
    • (2013) Pharmacol Rev , vol.65 , Issue.4 , pp. 1148-1161
    • Quintana, F.J.1    Sherr, D.H.2
  • 43
    • 84951866819 scopus 로고    scopus 로고
    • Aryl hydrocarbon receptor: Linking environment to immunity
    • Cella M, Colonna M (2015) Aryl hydrocarbon receptor: Linking environment to immunity. Semin Immunol 27(5):310-314.
    • (2015) Semin Immunol , vol.27 , Issue.5 , pp. 310-314
    • Cella, M.1    Colonna, M.2
  • 44
    • 84952864852 scopus 로고    scopus 로고
    • AHR function in lymphocytes: Emerging concepts
    • Zhou L (2016) AHR function in lymphocytes: Emerging concepts. Trends Immunol 37(1):17-31.
    • (2016) Trends Immunol , vol.37 , Issue.1 , pp. 17-31
    • Zhou, L.1
  • 45
    • 84950146989 scopus 로고    scopus 로고
    • Ultraviolet light converts propranolol, a nonselective β-blocker and potential lupus-inducing drug, into a proinflammatory AhR ligand
    • Dorgham K, et al. (2015) Ultraviolet light converts propranolol, a nonselective β-blocker and potential lupus-inducing drug, into a proinflammatory AhR ligand. Eur J Immunol 45(11):3174-3187.
    • (2015) Eur J Immunol , vol.45 , Issue.11 , pp. 3174-3187
    • Dorgham, K.1
  • 46
    • 84966526506 scopus 로고    scopus 로고
    • CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands
    • Lamas B, et al. (2016) CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med 22(6):598-605.
    • (2016) Nat Med , vol.22 , Issue.6 , pp. 598-605
    • Lamas, B.1
  • 47
    • 84871809302 scopus 로고    scopus 로고
    • STAR: Ultrafast universal RNA-seq aligner
    • Dobin A, et al. (2013) STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 29(1):15-21.
    • (2013) Bioinformatics , vol.29 , Issue.1 , pp. 15-21
    • Dobin, A.1
  • 48
    • 84897397058 scopus 로고    scopus 로고
    • FeatureCounts: An efficient general purpose program for assigning sequence reads to genomic features
    • Liao Y, Smyth GK, Shi W (2014) featureCounts: An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30(7):923-930.
    • (2014) Bioinformatics , vol.30 , Issue.7 , pp. 923-930
    • Liao, Y.1    Smyth, G.K.2    Shi, W.3
  • 49
    • 84865760395 scopus 로고    scopus 로고
    • GENCODE: The reference human genome annotation for The ENCODE Project
    • Harrow J, et al. (2012) GENCODE: The reference human genome annotation for The ENCODE Project. Genome Res 22(9):1760-1774.
    • (2012) Genome Res , vol.22 , Issue.9 , pp. 1760-1774
    • Harrow, J.1
  • 50
    • 84896735766 scopus 로고    scopus 로고
    • Voom: Precision weights unlock linear model analysis tools for RNA-seq read counts
    • Law CW, Chen Y, Shi W, Smyth GK (2014) voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol 15(2):R29.
    • (2014) Genome Biol , vol.15 , Issue.2 , pp. R29
    • Law, C.W.1    Chen, Y.2    Shi, W.3    Smyth, G.K.4
  • 51
    • 84926507971 scopus 로고    scopus 로고
    • Limma powers differential expression analyses for RNA-sequencing and microarray studies
    • Ritchie ME, et al. (2015) limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43(7):e47.
    • (2015) Nucleic Acids Res , vol.43 , Issue.7 , pp. e47
    • Ritchie, M.E.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.