Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2023

Open Access 01-12-2023 | Multiple Sclerosis | Research

Teriflunomide as a therapeutic means for myelin repair

Authors: Peter Göttle, Janos Groh, Laura Reiche, Joel Gruchot, Nicole Rychlik, Luisa Werner, Iria Samper Agrelo, Rainer Akkermann, Annika Zink, Alessandro Prigione, Hans-Peter Hartung, Rudolf Martini, Patrick Küry

Published in: Journal of Neuroinflammation | Issue 1/2023

Login to get access

Abstract

Background

Promotion of myelin repair in the context of demyelinating diseases such as multiple sclerosis (MS) still represents a clinical unmet need, given that this disease is not only characterized by autoimmune activities but also by impaired regeneration processes. Hence, this relates to replacement of lost oligodendrocytes and myelin sheaths—the primary targets of autoimmune attacks. Endogenous remyelination is mainly mediated via activation and differentiation of resident oligodendroglial precursor cells (OPCs), whereas its efficiency remains limited and declines with disease progression and aging. Teriflunomide has been approved as a first-line treatment for relapsing remitting MS. Beyond its role in acting via inhibition of de novo pyrimidine synthesis leading to a cytostatic effect on proliferating lymphocyte subsets, this study aims to uncover its potential to foster myelin repair.

Methods

Within the cuprizone mediated de-/remyelination model teriflunomide dependent effects on oligodendroglial homeostasis and maturation, related to cellular processes important for myelin repair were analyzed in vivo. Teriflunomide administration was performed either as pulse or continuously and markers specific for oligodendroglial maturation and mitochondrial integrity were examined by means of gene expression and immunohistochemical analyses. In addition, axon myelination was determined using electron microscopy.

Results

Both pulse and constant teriflunomide treatment efficiently boosted myelin repair activities in this model, leading to accelerated generation of oligodendrocytes and restoration of myelin sheaths. Moreover, teriflunomide restored mitochondrial integrity within oligodendroglial cells.

Conclusions

The link between de novo pyrimidine synthesis inhibition, oligodendroglial rescue, and maintenance of mitochondrial homeostasis appears as a key for successful myelin repair and hence for protection of axons from degeneration.
Literature
1.
go back to reference Lassmann H. Multiple sclerosis pathology. Cold Spring Harb Perspect Med. 2018;8(3): a028936.CrossRef Lassmann H. Multiple sclerosis pathology. Cold Spring Harb Perspect Med. 2018;8(3): a028936.CrossRef
2.
go back to reference Franklin RJ, Ffrench-Constant C. Remyelination in the CNS: from biology to therapy. Nat Rev Neurosci. 2008;9(11):839–55.CrossRef Franklin RJ, Ffrench-Constant C. Remyelination in the CNS: from biology to therapy. Nat Rev Neurosci. 2008;9(11):839–55.CrossRef
3.
go back to reference Zawadzka M, Rivers LE, Fancy SP, Zhao C, Tripathi R, Jamen F, et al. CNS-resident glial progenitor/stem cells produce Schwann cells as well as oligodendrocytes during repair of CNS demyelination. Cell Stem Cell. 2010;6(6):578–90.CrossRef Zawadzka M, Rivers LE, Fancy SP, Zhao C, Tripathi R, Jamen F, et al. CNS-resident glial progenitor/stem cells produce Schwann cells as well as oligodendrocytes during repair of CNS demyelination. Cell Stem Cell. 2010;6(6):578–90.CrossRef
4.
go back to reference Gensert JM, Goldman JE. Endogenous progenitors remyelinate demyelinated axons in the adult CNS. Neuron. 1997;19(1):197–203.CrossRef Gensert JM, Goldman JE. Endogenous progenitors remyelinate demyelinated axons in the adult CNS. Neuron. 1997;19(1):197–203.CrossRef
5.
go back to reference Simons M, Nave KA. Oligodendrocytes: myelination and axonal support. Cold Spring Harb Perspect Biol. 2015;8(1): a020479.CrossRef Simons M, Nave KA. Oligodendrocytes: myelination and axonal support. Cold Spring Harb Perspect Biol. 2015;8(1): a020479.CrossRef
6.
go back to reference Bezukladova S, Genchi A, Panina-Bordignon P, Martino G. Promoting exogenous repair in multiple sclerosis: myelin regeneration. Curr Opin Neurol. 2022;35(3):313–8.CrossRef Bezukladova S, Genchi A, Panina-Bordignon P, Martino G. Promoting exogenous repair in multiple sclerosis: myelin regeneration. Curr Opin Neurol. 2022;35(3):313–8.CrossRef
7.
go back to reference Lubetzki C, Zalc B, Kremer D, Küry P. Endogenous clues promoting remyelination in multiple sclerosis. Curr Opin Neurol. 2022;35(3):307–12.CrossRef Lubetzki C, Zalc B, Kremer D, Küry P. Endogenous clues promoting remyelination in multiple sclerosis. Curr Opin Neurol. 2022;35(3):307–12.CrossRef
8.
go back to reference Manousi A, Küry P. Small molecule screening as an approach to encounter inefficient myelin repair. Curr Opin Pharmacol. 2021;61:127–35.CrossRef Manousi A, Küry P. Small molecule screening as an approach to encounter inefficient myelin repair. Curr Opin Pharmacol. 2021;61:127–35.CrossRef
9.
go back to reference Küry P, Kremer D, Göttle P. Drug repurposing for neuroregeneration in multiple sclerosis. Neural Regen Res. 2018;13(8):1366–7.CrossRef Küry P, Kremer D, Göttle P. Drug repurposing for neuroregeneration in multiple sclerosis. Neural Regen Res. 2018;13(8):1366–7.CrossRef
10.
go back to reference Claussen MC, Korn T. Immune mechanisms of new therapeutic strategies in MS: teriflunomide. Clin Immunol. 2012;142(1):49–56.CrossRef Claussen MC, Korn T. Immune mechanisms of new therapeutic strategies in MS: teriflunomide. Clin Immunol. 2012;142(1):49–56.CrossRef
11.
go back to reference O’Connor P, Wolinsky JS, Confavreux C, Comi G, Kappos L, Olsson TP, et al. Randomized trial of oral teriflunomide for relapsing multiple sclerosis. N Engl J Med. 2011;365(14):1293–303.CrossRef O’Connor P, Wolinsky JS, Confavreux C, Comi G, Kappos L, Olsson TP, et al. Randomized trial of oral teriflunomide for relapsing multiple sclerosis. N Engl J Med. 2011;365(14):1293–303.CrossRef
12.
go back to reference Confavreux C, O’Connor P, Comi G, Freedman MS, Miller AE, Olsson TP, et al. Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol. 2014;13(3):247–56.CrossRef Confavreux C, O’Connor P, Comi G, Freedman MS, Miller AE, Olsson TP, et al. Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol. 2014;13(3):247–56.CrossRef
13.
go back to reference Bruneau JM, Yea CM, Spinella-Jaegle S, Fudali C, Woodward K, Robson PA, et al. Purification of human dihydro-orotate dehydrogenase and its inhibition by A77 1726, the active metabolite of leflunomide. Biochem J. 1998;336(Pt 2):299–303.CrossRef Bruneau JM, Yea CM, Spinella-Jaegle S, Fudali C, Woodward K, Robson PA, et al. Purification of human dihydro-orotate dehydrogenase and its inhibition by A77 1726, the active metabolite of leflunomide. Biochem J. 1998;336(Pt 2):299–303.CrossRef
14.
go back to reference Cherwinski HM, Cohn RG, Cheung P, Webster DJ, Xu YZ, Caulfield JP, et al. The immunosuppressant leflunomide inhibits lymphocyte proliferation by inhibiting pyrimidine biosynthesis. J Pharmacol Exp Ther. 1995;275(2):1043–9. Cherwinski HM, Cohn RG, Cheung P, Webster DJ, Xu YZ, Caulfield JP, et al. The immunosuppressant leflunomide inhibits lymphocyte proliferation by inhibiting pyrimidine biosynthesis. J Pharmacol Exp Ther. 1995;275(2):1043–9.
15.
go back to reference Zhang J, Teran G, Popa M, Madapura H, Ladds M, Lianoudaki D, et al. DHODH inhibition modulates glucose metabolism and circulating GDF15, and improves metabolic balance. iScience. 2021;24(5): 102494.CrossRef Zhang J, Teran G, Popa M, Madapura H, Ladds M, Lianoudaki D, et al. DHODH inhibition modulates glucose metabolism and circulating GDF15, and improves metabolic balance. iScience. 2021;24(5): 102494.CrossRef
16.
go back to reference Göttle P, Manousi A, Kremer D, Reiche L, Hartung HP, Küry P. Teriflunomide promotes oligodendroglial differentiation and myelination. J Neuroinflammation. 2018;15(1):76.CrossRef Göttle P, Manousi A, Kremer D, Reiche L, Hartung HP, Küry P. Teriflunomide promotes oligodendroglial differentiation and myelination. J Neuroinflammation. 2018;15(1):76.CrossRef
18.
go back to reference Matsushima GK, Morell P. The neurotoxicant, cuprizone, as a model to study demyelination and remyelination in the central nervous system. Brain Pathol. 2001;11(1):107–16.CrossRef Matsushima GK, Morell P. The neurotoxicant, cuprizone, as a model to study demyelination and remyelination in the central nervous system. Brain Pathol. 2001;11(1):107–16.CrossRef
19.
go back to reference Ringheim GE, Lee L, Laws-Ricker L, Delohery T, Liu L, Zhang D, et al. Teriflunomide attenuates immunopathological changes in the dark agouti rat model of experimental autoimmune encephalomyelitis. Front Neurol. 2013;4:169.CrossRef Ringheim GE, Lee L, Laws-Ricker L, Delohery T, Liu L, Zhang D, et al. Teriflunomide attenuates immunopathological changes in the dark agouti rat model of experimental autoimmune encephalomyelitis. Front Neurol. 2013;4:169.CrossRef
20.
go back to reference Groh J, Horner M, Martini R. Teriflunomide attenuates neuroinflammation-related neural damage in mice carrying human PLP1 mutations. J Neuroinflammation. 2018;15(1):194.CrossRef Groh J, Horner M, Martini R. Teriflunomide attenuates neuroinflammation-related neural damage in mice carrying human PLP1 mutations. J Neuroinflammation. 2018;15(1):194.CrossRef
21.
go back to reference Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016;7(2):27–31.CrossRef Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016;7(2):27–31.CrossRef
22.
go back to reference Chen Z, Chen JT, Johnson M, Gossman ZC, Hendrickson M, Sakaie K, et al. Cuprizone does not induce CNS demyelination in nonhuman primates. Ann Clin Transl Neurol. 2015;2(2):208–13.CrossRef Chen Z, Chen JT, Johnson M, Gossman ZC, Hendrickson M, Sakaie K, et al. Cuprizone does not induce CNS demyelination in nonhuman primates. Ann Clin Transl Neurol. 2015;2(2):208–13.CrossRef
23.
go back to reference Nakatani H, Martin E, Hassani H, Clavairoly A, Maire CL, Viadieu A, et al. Ascl1/Mash1 promotes brain oligodendrogenesis during myelination and remyelination. J Neurosci. 2013;33(23):9752–68.CrossRef Nakatani H, Martin E, Hassani H, Clavairoly A, Maire CL, Viadieu A, et al. Ascl1/Mash1 promotes brain oligodendrogenesis during myelination and remyelination. J Neurosci. 2013;33(23):9752–68.CrossRef
24.
go back to reference Göttle P, Sabo JK, Heinen A, Venables G, Torres K, Tzekova N, et al. Oligodendroglial maturation is dependent on intracellular protein shuttling. J Neurosci. 2015;35(3):906–19.CrossRef Göttle P, Sabo JK, Heinen A, Venables G, Torres K, Tzekova N, et al. Oligodendroglial maturation is dependent on intracellular protein shuttling. J Neurosci. 2015;35(3):906–19.CrossRef
25.
go back to reference Pavic G, Petzsch P, Jansen R, Raba K, Rychlik N, Simiantonakis I, et al. Microglia contributes to remyelination in cerebral but not spinal cord ischemia. Glia. 2021;69(11):2739–51.CrossRef Pavic G, Petzsch P, Jansen R, Raba K, Rychlik N, Simiantonakis I, et al. Microglia contributes to remyelination in cerebral but not spinal cord ischemia. Glia. 2021;69(11):2739–51.CrossRef
26.
go back to reference Groh J, Friedman HC, Orel N, Ip CW, Fischer S, Spahn I, et al. Pathogenic inflammation in the CNS of mice carrying human PLP1 mutations. Hum Mol Genet. 2016;25(21):4686–702. Groh J, Friedman HC, Orel N, Ip CW, Fischer S, Spahn I, et al. Pathogenic inflammation in the CNS of mice carrying human PLP1 mutations. Hum Mol Genet. 2016;25(21):4686–702.
27.
go back to reference Chaudhry A, Shi R, Luciani DS. A pipeline for multidimensional confocal analysis of mitochondrial morphology, function, and dynamics in pancreatic beta-cells. Am J Physiol Endocrinol Metab. 2020;318(2):E87–101.CrossRef Chaudhry A, Shi R, Luciani DS. A pipeline for multidimensional confocal analysis of mitochondrial morphology, function, and dynamics in pancreatic beta-cells. Am J Physiol Endocrinol Metab. 2020;318(2):E87–101.CrossRef
28.
go back to reference Hemel I, Engelen BPH, Luber N, Gerards M. A hitchhiker’s guide to mitochondrial quantification. Mitochondrion. 2021;59:216–24.CrossRef Hemel I, Engelen BPH, Luber N, Gerards M. A hitchhiker’s guide to mitochondrial quantification. Mitochondrion. 2021;59:216–24.CrossRef
29.
go back to reference Faul F, Erdfelder E, Buchner A, Lang AG. Statistical power analyses using G*Power 3.1: tests for correlation and regression analyses. Behav Res Methods. 2009;41(4):1149–60.CrossRef Faul F, Erdfelder E, Buchner A, Lang AG. Statistical power analyses using G*Power 3.1: tests for correlation and regression analyses. Behav Res Methods. 2009;41(4):1149–60.CrossRef
30.
go back to reference Xing YL, Roth PT, Stratton JA, Chuang BH, Danne J, Ellis SL, et al. Adult neural precursor cells from the subventricular zone contribute significantly to oligodendrocyte regeneration and remyelination. J Neurosci. 2014;34(42):14128–46.CrossRef Xing YL, Roth PT, Stratton JA, Chuang BH, Danne J, Ellis SL, et al. Adult neural precursor cells from the subventricular zone contribute significantly to oligodendrocyte regeneration and remyelination. J Neurosci. 2014;34(42):14128–46.CrossRef
31.
go back to reference Bujalka H, Koenning M, Jackson S, Perreau VM, Pope B, Hay CM, et al. MYRF is a membrane-associated transcription factor that autoproteolytically cleaves to directly activate myelin genes. PLoS Biol. 2013;11(8): e1001625.CrossRef Bujalka H, Koenning M, Jackson S, Perreau VM, Pope B, Hay CM, et al. MYRF is a membrane-associated transcription factor that autoproteolytically cleaves to directly activate myelin genes. PLoS Biol. 2013;11(8): e1001625.CrossRef
32.
go back to reference Spaas J, van Veggel L, Schepers M, Tiane A, van Horssen J, Wilson DM 3rd, et al. Oxidative stress and impaired oligodendrocyte precursor cell differentiation in neurological disorders. Cell Mol Life Sci. 2021;78(10):4615–37.CrossRef Spaas J, van Veggel L, Schepers M, Tiane A, van Horssen J, Wilson DM 3rd, et al. Oxidative stress and impaired oligodendrocyte precursor cell differentiation in neurological disorders. Cell Mol Life Sci. 2021;78(10):4615–37.CrossRef
33.
go back to reference Corona JC, Duchen MR. PPARgamma as a therapeutic target to rescue mitochondrial function in neurological disease. Free Radic Biol Med. 2016;100:153–63.CrossRef Corona JC, Duchen MR. PPARgamma as a therapeutic target to rescue mitochondrial function in neurological disease. Free Radic Biol Med. 2016;100:153–63.CrossRef
34.
go back to reference Yeligar SM, Kang BY, Bijli KM, Kleinhenz JM, Murphy TC, Torres G, et al. PPARgamma regulates mitochondrial structure and function and human pulmonary artery smooth muscle cell proliferation. Am J Respir Cell Mol Biol. 2018;58(5):648–57.CrossRef Yeligar SM, Kang BY, Bijli KM, Kleinhenz JM, Murphy TC, Torres G, et al. PPARgamma regulates mitochondrial structure and function and human pulmonary artery smooth muscle cell proliferation. Am J Respir Cell Mol Biol. 2018;58(5):648–57.CrossRef
35.
go back to reference Sidarala V, Zhu J, Levi-D’Ancona E, Pearson GL, Reck EC, Walker EM, et al. Mitofusin 1 and 2 regulation of mitochondrial DNA content is a critical determinant of glucose homeostasis. Nat Commun. 2022;13(1):2340.CrossRef Sidarala V, Zhu J, Levi-D’Ancona E, Pearson GL, Reck EC, Walker EM, et al. Mitofusin 1 and 2 regulation of mitochondrial DNA content is a critical determinant of glucose homeostasis. Nat Commun. 2022;13(1):2340.CrossRef
36.
go back to reference Gao F, Reynolds MB, Passalacqua KD, Sexton JZ, Abuaita BH, O’Riordan MXD. The mitochondrial fission regulator DRP1 controls post-transcriptional regulation of TNF-alpha. Front Cell Infect Microbiol. 2020;10: 593805.CrossRef Gao F, Reynolds MB, Passalacqua KD, Sexton JZ, Abuaita BH, O’Riordan MXD. The mitochondrial fission regulator DRP1 controls post-transcriptional regulation of TNF-alpha. Front Cell Infect Microbiol. 2020;10: 593805.CrossRef
37.
go back to reference Lisowski P, Kannan P, Mlody B, Prigione A. Mitochondria and the dynamic control of stem cell homeostasis. EMBO Rep. 2018;19(5):e45432.CrossRef Lisowski P, Kannan P, Mlody B, Prigione A. Mitochondria and the dynamic control of stem cell homeostasis. EMBO Rep. 2018;19(5):e45432.CrossRef
38.
go back to reference Peng F, Wang JH, Fan WJ, Meng YT, Li MM, Li TT, et al. Glycolysis gatekeeper PDK1 reprograms breast cancer stem cells under hypoxia. Oncogene. 2018;37(8):1119.CrossRef Peng F, Wang JH, Fan WJ, Meng YT, Li MM, Li TT, et al. Glycolysis gatekeeper PDK1 reprograms breast cancer stem cells under hypoxia. Oncogene. 2018;37(8):1119.CrossRef
39.
go back to reference Xie H, Hanai J, Ren JG, Kats L, Burgess K, Bhargava P, et al. Targeting lactate dehydrogenase–a inhibits tumorigenesis and tumor progression in mouse models of lung cancer and impacts tumor-initiating cells. Cell Metab. 2014;19(5):795–809.CrossRef Xie H, Hanai J, Ren JG, Kats L, Burgess K, Bhargava P, et al. Targeting lactate dehydrogenase–a inhibits tumorigenesis and tumor progression in mouse models of lung cancer and impacts tumor-initiating cells. Cell Metab. 2014;19(5):795–809.CrossRef
40.
go back to reference Dupuy F, Tabaries S, Andrzejewski S, Dong Z, Blagih J, Annis MG, et al. PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer. Cell Metab. 2015;22(4):577–89.CrossRef Dupuy F, Tabaries S, Andrzejewski S, Dong Z, Blagih J, Annis MG, et al. PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer. Cell Metab. 2015;22(4):577–89.CrossRef
41.
go back to reference Prigione A, Rohwer N, Hoffmann S, Mlody B, Drews K, Bukowiecki R, et al. HIF1alpha modulates cell fate reprogramming through early glycolytic shift and upregulation of PDK1-3 and PKM2. Stem Cells. 2014;32(2):364–76.CrossRef Prigione A, Rohwer N, Hoffmann S, Mlody B, Drews K, Bukowiecki R, et al. HIF1alpha modulates cell fate reprogramming through early glycolytic shift and upregulation of PDK1-3 and PKM2. Stem Cells. 2014;32(2):364–76.CrossRef
42.
go back to reference Bar-Or A, Pachner A, Menguy-Vacheron F, Kaplan J, Wiendl H. Teriflunomide and its mechanism of action in multiple sclerosis. Drugs. 2014;74(6):659–74.CrossRef Bar-Or A, Pachner A, Menguy-Vacheron F, Kaplan J, Wiendl H. Teriflunomide and its mechanism of action in multiple sclerosis. Drugs. 2014;74(6):659–74.CrossRef
43.
go back to reference Lu Z, Zhang D, Cui K, Fu X, Man J, Lu H, et al. Neuroprotective action of teriflunomide in a mouse model of transient middle cerebral artery occlusion. Neuroscience. 2020;428:228–41.CrossRef Lu Z, Zhang D, Cui K, Fu X, Man J, Lu H, et al. Neuroprotective action of teriflunomide in a mouse model of transient middle cerebral artery occlusion. Neuroscience. 2020;428:228–41.CrossRef
44.
go back to reference Freedman MS, Wolinsky JS, Comi G, Kappos L, Olsson TP, Miller AE, et al. The efficacy of teriflunomide in patients who received prior disease-modifying treatments: subgroup analyses of the teriflunomide phase 3 TEMSO and TOWER studies. Mult Scler. 2018;24(4):535–9.CrossRef Freedman MS, Wolinsky JS, Comi G, Kappos L, Olsson TP, Miller AE, et al. The efficacy of teriflunomide in patients who received prior disease-modifying treatments: subgroup analyses of the teriflunomide phase 3 TEMSO and TOWER studies. Mult Scler. 2018;24(4):535–9.CrossRef
45.
go back to reference Barcelos IP, Troxell RM, Graves JS. Mitochondrial dysfunction and multiple sclerosis. Biology (Basel). 2019;8(2):37. Barcelos IP, Troxell RM, Graves JS. Mitochondrial dysfunction and multiple sclerosis. Biology (Basel). 2019;8(2):37.
46.
go back to reference Zhao JW, Wang DX, Ma XR, Dong ZJ, Wu JB, Wang F, et al. Impaired metabolism of oligodendrocyte progenitor cells and axons in demyelinated lesion and in the aged CNS. Curr Opin Pharmacol. 2022;64: 102205.CrossRef Zhao JW, Wang DX, Ma XR, Dong ZJ, Wu JB, Wang F, et al. Impaired metabolism of oligodendrocyte progenitor cells and axons in demyelinated lesion and in the aged CNS. Curr Opin Pharmacol. 2022;64: 102205.CrossRef
47.
go back to reference Rinholm JE, Vervaeke K, Tadross MR, Tkachuk AN, Kopek BG, Brown TA, et al. Movement and structure of mitochondria in oligodendrocytes and their myelin sheaths. Glia. 2016;64(5):810–25.CrossRef Rinholm JE, Vervaeke K, Tadross MR, Tkachuk AN, Kopek BG, Brown TA, et al. Movement and structure of mitochondria in oligodendrocytes and their myelin sheaths. Glia. 2016;64(5):810–25.CrossRef
48.
go back to reference Bertholet AM, Delerue T, Millet AM, Moulis MF, David C, Daloyau M, et al. Mitochondrial fusion/fission dynamics in neurodegeneration and neuronal plasticity. Neurobiol Dis. 2016;90:3–19.CrossRef Bertholet AM, Delerue T, Millet AM, Moulis MF, David C, Daloyau M, et al. Mitochondrial fusion/fission dynamics in neurodegeneration and neuronal plasticity. Neurobiol Dis. 2016;90:3–19.CrossRef
49.
go back to reference Meyer N, Rinholm JE. Mitochondria in myelinating oligodendrocytes: slow and out of breath? Metabolites. 2021;11(6):359.CrossRef Meyer N, Rinholm JE. Mitochondria in myelinating oligodendrocytes: slow and out of breath? Metabolites. 2021;11(6):359.CrossRef
50.
go back to reference Marangon D, Boccazzi M, Lecca D, Fumagalli M. Regulation of oligodendrocyte functions: targeting lipid metabolism and extracellular matrix for myelin repair. J Clin Med. 2020;9(2):470.CrossRef Marangon D, Boccazzi M, Lecca D, Fumagalli M. Regulation of oligodendrocyte functions: targeting lipid metabolism and extracellular matrix for myelin repair. J Clin Med. 2020;9(2):470.CrossRef
51.
go back to reference Poitelon Y, Kopec AM, Belin S. Myelin fat facts: an overview of lipids and fatty acid metabolism. Cells. 2020;9(4):812.CrossRef Poitelon Y, Kopec AM, Belin S. Myelin fat facts: an overview of lipids and fatty acid metabolism. Cells. 2020;9(4):812.CrossRef
52.
go back to reference Tepavcevic V. Oligodendroglial energy metabolism and (re)myelination. Life. 2021;11(3):238.CrossRef Tepavcevic V. Oligodendroglial energy metabolism and (re)myelination. Life. 2021;11(3):238.CrossRef
53.
go back to reference Schoenfeld R, Wong A, Silva J, Li M, Itoh A, Horiuchi M, et al. Oligodendroglial differentiation induces mitochondrial genes and inhibition of mitochondrial function represses oligodendroglial differentiation. Mitochondrion. 2010;10(2):143–50.CrossRef Schoenfeld R, Wong A, Silva J, Li M, Itoh A, Horiuchi M, et al. Oligodendroglial differentiation induces mitochondrial genes and inhibition of mitochondrial function represses oligodendroglial differentiation. Mitochondrion. 2010;10(2):143–50.CrossRef
54.
go back to reference Yazdankhah M, Shang P, Ghosh S, Bhutto IA, Stepicheva N, Grebe R, et al. Modulating EGFR-MTORC1-autophagy as a potential therapy for persistent fetal vasculature (PFV) disease. Autophagy. 2020;16(6):1130–42.CrossRef Yazdankhah M, Shang P, Ghosh S, Bhutto IA, Stepicheva N, Grebe R, et al. Modulating EGFR-MTORC1-autophagy as a potential therapy for persistent fetal vasculature (PFV) disease. Autophagy. 2020;16(6):1130–42.CrossRef
55.
go back to reference Tondera D, Grandemange S, Jourdain A, Karbowski M, Mattenberger Y, Herzig S, et al. SLP-2 is required for stress-induced mitochondrial hyperfusion. EMBO J. 2009;28(11):1589–600.CrossRef Tondera D, Grandemange S, Jourdain A, Karbowski M, Mattenberger Y, Herzig S, et al. SLP-2 is required for stress-induced mitochondrial hyperfusion. EMBO J. 2009;28(11):1589–600.CrossRef
56.
go back to reference Mari M, Colell A. Mitochondrial oxidative and nitrosative stress as a therapeutic target in diseases. Antioxidants. 2021;10(2):314.CrossRef Mari M, Colell A. Mitochondrial oxidative and nitrosative stress as a therapeutic target in diseases. Antioxidants. 2021;10(2):314.CrossRef
57.
go back to reference Rojo M, Legros F, Chateau D, Lombes A. Membrane topology and mitochondrial targeting of mitofusins, ubiquitous mammalian homologs of the transmembrane GTPase Fzo. J Cell Sci. 2002;115(Pt 8):1663–74.CrossRef Rojo M, Legros F, Chateau D, Lombes A. Membrane topology and mitochondrial targeting of mitofusins, ubiquitous mammalian homologs of the transmembrane GTPase Fzo. J Cell Sci. 2002;115(Pt 8):1663–74.CrossRef
58.
go back to reference Malla B, Liotta A, Bros H, Ulshofer R, Paul F, Hauser AE, et al. Teriflunomide preserves neuronal activity and protects mitochondria in brain slices exposed to oxidative stress. Int J Mol Sci. 2022;23(3):1538.CrossRef Malla B, Liotta A, Bros H, Ulshofer R, Paul F, Hauser AE, et al. Teriflunomide preserves neuronal activity and protects mitochondria in brain slices exposed to oxidative stress. Int J Mol Sci. 2022;23(3):1538.CrossRef
59.
go back to reference Miret-Casals L, Sebastian D, Brea J, Rico-Leo EM, Palacin M, Fernandez-Salguero PM, et al. Identification of new activators of mitochondrial fusion reveals a link between mitochondrial morphology and pyrimidine metabolism. Cell Chem Biol. 2018;25(3):268-78 e4.CrossRef Miret-Casals L, Sebastian D, Brea J, Rico-Leo EM, Palacin M, Fernandez-Salguero PM, et al. Identification of new activators of mitochondrial fusion reveals a link between mitochondrial morphology and pyrimidine metabolism. Cell Chem Biol. 2018;25(3):268-78 e4.CrossRef
60.
go back to reference Pellattiero A, Scorrano L. Flaming mitochondria: the anti-inflammatory drug leflunomide boosts mitofusins. Cell Chem Biol. 2018;25(3):231–3.CrossRef Pellattiero A, Scorrano L. Flaming mitochondria: the anti-inflammatory drug leflunomide boosts mitofusins. Cell Chem Biol. 2018;25(3):231–3.CrossRef
61.
go back to reference Magalon K, Le Grand M, El Waly B, Moulis M, Pruss R, Bordet T, et al. Olesoxime favors oligodendrocyte differentiation through a functional interplay between mitochondria and microtubules. Neuropharmacology. 2016;111:293–303.CrossRef Magalon K, Le Grand M, El Waly B, Moulis M, Pruss R, Bordet T, et al. Olesoxime favors oligodendrocyte differentiation through a functional interplay between mitochondria and microtubules. Neuropharmacology. 2016;111:293–303.CrossRef
62.
go back to reference Iwata K, Scorrano L. Finding a new balance to cure Charcot-Marie-Tooth 2A. J Clin Invest. 2019;129(4):1533–5.CrossRef Iwata K, Scorrano L. Finding a new balance to cure Charcot-Marie-Tooth 2A. J Clin Invest. 2019;129(4):1533–5.CrossRef
63.
go back to reference Pfeuffer S, Kerschke L, Ruck T, Rolfes L, Pawlitzki M, Albrecht P, et al. Teriflunomide treatment is associated with optic nerve recovery in early multiple sclerosis. Ther Adv Neurol Disord. 2021;14:1756286421997372.CrossRef Pfeuffer S, Kerschke L, Ruck T, Rolfes L, Pawlitzki M, Albrecht P, et al. Teriflunomide treatment is associated with optic nerve recovery in early multiple sclerosis. Ther Adv Neurol Disord. 2021;14:1756286421997372.CrossRef
64.
go back to reference Zhan J, Mann T, Joost S, Behrangi N, Frank M, Kipp M. The cuprizone model: dos and do nots. Cells. 2020;9(4):843.CrossRef Zhan J, Mann T, Joost S, Behrangi N, Frank M, Kipp M. The cuprizone model: dos and do nots. Cells. 2020;9(4):843.CrossRef
65.
go back to reference Kabiraj P, Grund EM, Clarkson BDS, Johnson RK, LaFrance-Corey RG, Lucchinetti CF, et al. Teriflunomide shifts the astrocytic bioenergetic profile from oxidative metabolism to glycolysis and attenuates TNFalpha-induced inflammatory responses. Sci Rep. 2022;12(1):3049.CrossRef Kabiraj P, Grund EM, Clarkson BDS, Johnson RK, LaFrance-Corey RG, Lucchinetti CF, et al. Teriflunomide shifts the astrocytic bioenergetic profile from oxidative metabolism to glycolysis and attenuates TNFalpha-induced inflammatory responses. Sci Rep. 2022;12(1):3049.CrossRef
66.
go back to reference Silva Oliveira M, Schira-Heinen J, Reiche L, Han S, de Amorim VCM, Lewen I, et al. Myelin repair is fostered by the corticosteroid medrysone specifically acting on astroglial subpopulations. EBioMedicine. 2022;83:104204.CrossRef Silva Oliveira M, Schira-Heinen J, Reiche L, Han S, de Amorim VCM, Lewen I, et al. Myelin repair is fostered by the corticosteroid medrysone specifically acting on astroglial subpopulations. EBioMedicine. 2022;83:104204.CrossRef
Metadata
Title
Teriflunomide as a therapeutic means for myelin repair
Authors
Peter Göttle
Janos Groh
Laura Reiche
Joel Gruchot
Nicole Rychlik
Luisa Werner
Iria Samper Agrelo
Rainer Akkermann
Annika Zink
Alessandro Prigione
Hans-Peter Hartung
Rudolf Martini
Patrick Küry
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2023
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-022-02686-6

Other articles of this Issue 1/2023

Journal of Neuroinflammation 1/2023 Go to the issue