Skip to main content
Top
Published in: NeuroMolecular Medicine 3/2019

01-09-2019 | Alzheimer's Disease | Review Paper

Beneficial Effects of Fingolimod in Alzheimer’s Disease: Molecular Mechanisms and Therapeutic Potential

Authors: Efthalia Angelopoulou, Christina Piperi

Published in: NeuroMolecular Medicine | Issue 3/2019

Login to get access

Abstract

Alzheimer’s disease (AD), the most common cause of dementia remains of unclear etiology with current pharmacological therapies failing to halt disease progression. Several pathophysiological mechanisms have been implicated in AD pathogenesis including amyloid-β protein (Aβ) accumulation, tau hyperphosphorylation, neuroinflammation and alterations in bioactive lipid metabolism. Sphingolipids, such as sphingosine-1-phosphate (S1P) and intracellular ceramide/S1P balance are highly implicated in central nervous system physiology as well as in AD pathogenesis. FTY720/Fingolimod, a structural sphingosine analog and S1P receptor (S1PR) modulator that is currently used in the treatment of relapsing–remitting multiple sclerosis (RRMS) has been shown to exert beneficial effects on AD progression. Recent in vitro and in vivo evidence indicate that fingolimod may suppress Aβ secretion and deposition, inhibit apoptosis and enhance brain-derived neurotrophic factor (BDNF) production. Furthermore, it regulates neuroinflammation, protects against N-methyl-d-aspartate (NMDA)-excitotoxicity and modulates receptor for advanced glycation end products signaling axis that is highly implicated in AD pathogenesis. This review discusses the underlying molecular mechanisms of the emerging neuroprotective role of fingolimod in AD and its therapeutic potential, aiming to shed more light on AD pathogenesis as well as direct future treatment strategies.
Literature
go back to reference Bateman, R. J., Aisen, P. S., De Strooper, B., Fox, N. C., Lemere, C. A., Ringman, J. M., et al. (2011). Autosomal-dominant Alzheimer’s disease: A review and proposal for the prevention of Alzheimer’s disease. Alzheimer’s Research & Therapy, 3(1), 1. https://doi.org/10.1186/alzrt59.CrossRef Bateman, R. J., Aisen, P. S., De Strooper, B., Fox, N. C., Lemere, C. A., Ringman, J. M., et al. (2011). Autosomal-dominant Alzheimer’s disease: A review and proposal for the prevention of Alzheimer’s disease. Alzheimer’s Research & Therapy, 3(1), 1. https://​doi.​org/​10.​1186/​alzrt59.CrossRef
go back to reference Bianca, V. D., Dusi, S., Bianchini, E., Dal Pra, I., & Rossi, F. (1999). Beta-amyloid activates the O-2 forming NADPH oxidase in microglia, monocytes, and neutrophils. A possible inflammatory mechanism of neuronal damage in Alzheimer’s disease. The Journal of Biological Chemistry, 274(22), 15493–15499. https://doi.org/10.1074/jbc.274.22.15493.CrossRefPubMed Bianca, V. D., Dusi, S., Bianchini, E., Dal Pra, I., & Rossi, F. (1999). Beta-amyloid activates the O-2 forming NADPH oxidase in microglia, monocytes, and neutrophils. A possible inflammatory mechanism of neuronal damage in Alzheimer’s disease. The Journal of Biological Chemistry, 274(22), 15493–15499. https://​doi.​org/​10.​1074/​jbc.​274.​22.​15493.CrossRefPubMed
go back to reference Brinkmann, V., Pinschewer, D. D., Feng, L., & Chen, S. (2001). FTY720: Altered lymphocyte traffic results in allograft protection. Transplantation, 72(5), 764–769.CrossRef Brinkmann, V., Pinschewer, D. D., Feng, L., & Chen, S. (2001). FTY720: Altered lymphocyte traffic results in allograft protection. Transplantation, 72(5), 764–769.CrossRef
go back to reference Choi, J. W., Gardell, S. E., Herr, D. R., Rivera, R., Lee, C. W., Noguchi, K., et al. (2011). FTY720 (fingolimod) efficacy in an animal model of multiple sclerosis requires astrocyte sphingosine 1-phosphate receptor 1 (S1P1) modulation. Proceedings of the National academy of Sciences of the United States of America, 108(2), 751–756. https://doi.org/10.1073/pnas.1014154108.CrossRefPubMed Choi, J. W., Gardell, S. E., Herr, D. R., Rivera, R., Lee, C. W., Noguchi, K., et al. (2011). FTY720 (fingolimod) efficacy in an animal model of multiple sclerosis requires astrocyte sphingosine 1-phosphate receptor 1 (S1P1) modulation. Proceedings of the National academy of Sciences of the United States of America, 108(2), 751–756. https://​doi.​org/​10.​1073/​pnas.​1014154108.CrossRefPubMed
go back to reference Clausznitzer, D., Pichardo-Almarza, C., Relo, A. L., van Bergeijk, J., van der Kam, E., Laplanche, L., et al. (2018). Quantitative systems pharmacology model for alzheimer disease indicates targeting sphingolipid dysregulation as potential treatment option. CPT Pharmacometrics & Systems Pharmacology, 7(11), 759–770. https://doi.org/10.1002/psp4.12351.CrossRef Clausznitzer, D., Pichardo-Almarza, C., Relo, A. L., van Bergeijk, J., van der Kam, E., Laplanche, L., et al. (2018). Quantitative systems pharmacology model for alzheimer disease indicates targeting sphingolipid dysregulation as potential treatment option. CPT Pharmacometrics & Systems Pharmacology, 7(11), 759–770. https://​doi.​org/​10.​1002/​psp4.​12351.CrossRef
go back to reference Cui, Y., Le, Y., Yazawa, H., Gong, W., & Wang, J. M. (2002). Potential role of the formyl peptide receptor-like 1 (FPRL1) in inflammatory aspects of Alzheimer’s disease. Journal of Leukocyte Biology, 72(4), 628–635.PubMed Cui, Y., Le, Y., Yazawa, H., Gong, W., & Wang, J. M. (2002). Potential role of the formyl peptide receptor-like 1 (FPRL1) in inflammatory aspects of Alzheimer’s disease. Journal of Leukocyte Biology, 72(4), 628–635.PubMed
go back to reference Cummings, J., Gould, H., & Zhong, K. (2012). Advances in designs for Alzheimer’s disease clinical trials. American Journal of Neurodegenerative Disease, 1(3), 205–216.PubMedPubMedCentral Cummings, J., Gould, H., & Zhong, K. (2012). Advances in designs for Alzheimer’s disease clinical trials. American Journal of Neurodegenerative Disease, 1(3), 205–216.PubMedPubMedCentral
go back to reference Czech, B., Pfeilschifter, W., Mazaheri-Omrani, N., Strobel, M. A., Kahles, T., Neumann-Haefelin, T., et al. (2009). The immunomodulatory sphingosine 1-phosphate analog FTY720 reduces lesion size and improves neurological outcome in a mouse model of cerebral ischemia. Biochemical and Biophysical Research Communications, 389(2), 251–256. https://doi.org/10.1016/j.bbrc.2009.08.142.CrossRefPubMed Czech, B., Pfeilschifter, W., Mazaheri-Omrani, N., Strobel, M. A., Kahles, T., Neumann-Haefelin, T., et al. (2009). The immunomodulatory sphingosine 1-phosphate analog FTY720 reduces lesion size and improves neurological outcome in a mouse model of cerebral ischemia. Biochemical and Biophysical Research Communications, 389(2), 251–256. https://​doi.​org/​10.​1016/​j.​bbrc.​2009.​08.​142.CrossRefPubMed
go back to reference Kaneider, N. C., Lindner, J., Feistritzer, C., Sturn, D. H., Mosheimer, B. A., Djanani, A. M., et al. (2004). The immune modulator FTY720 targets sphingosine-kinase-dependent migration of human monocytes in response to amyloid beta-protein and its precursor. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology, 18(11), 1309–1311. https://doi.org/10.1096/fj.03-1050fje.CrossRef Kaneider, N. C., Lindner, J., Feistritzer, C., Sturn, D. H., Mosheimer, B. A., Djanani, A. M., et al. (2004). The immune modulator FTY720 targets sphingosine-kinase-dependent migration of human monocytes in response to amyloid beta-protein and its precursor. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology, 18(11), 1309–1311. https://​doi.​org/​10.​1096/​fj.​03-1050fje.CrossRef
go back to reference Lahiri, S., Park, H., Laviad, E. L., Lu, X., Bittman, R., & Futerman, A. H. (2009). Ceramide synthesis is modulated by the sphingosine analog FTY720 via a mixture of uncompetitive and noncompetitive inhibition in an Acyl-CoA chain length-dependent manner. Journal of Biological Chemistry, 284(24), 16090–16098. https://doi.org/10.1074/jbc.M807438200.CrossRefPubMed Lahiri, S., Park, H., Laviad, E. L., Lu, X., Bittman, R., & Futerman, A. H. (2009). Ceramide synthesis is modulated by the sphingosine analog FTY720 via a mixture of uncompetitive and noncompetitive inhibition in an Acyl-CoA chain length-dependent manner. Journal of Biological Chemistry, 284(24), 16090–16098. https://​doi.​org/​10.​1074/​jbc.​M807438200.CrossRefPubMed
go back to reference Lorton, D. (1997). beta-Amyloid-induced IL-1 beta release from an activated human monocyte cell line is calcium- and G-protein-dependent. Mechanisms of Ageing and Development, 94(1–3), 199–211.CrossRef Lorton, D. (1997). beta-Amyloid-induced IL-1 beta release from an activated human monocyte cell line is calcium- and G-protein-dependent. Mechanisms of Ageing and Development, 94(1–3), 199–211.CrossRef
go back to reference McManus, R. M., Finucane, O. M., Wilk, M. M., Mills, K. H. G., & Lynch, M. A. (2017). FTY720 Attenuates Infection-Induced Enhancement of Aβ Accumulation in APP/PS1 Mice by Modulating Astrocytic Activation. Journal of Neuroimmune Pharmacology: The Official Journal of the Society on NeuroImmune Pharmacology, 12(4), 670–681. https://doi.org/10.1007/s11481-017-9753-6.CrossRef McManus, R. M., Finucane, O. M., Wilk, M. M., Mills, K. H. G., & Lynch, M. A. (2017). FTY720 Attenuates Infection-Induced Enhancement of Aβ Accumulation in APP/PS1 Mice by Modulating Astrocytic Activation. Journal of Neuroimmune Pharmacology: The Official Journal of the Society on NeuroImmune Pharmacology, 12(4), 670–681. https://​doi.​org/​10.​1007/​s11481-017-9753-6.CrossRef
go back to reference Mendell, J., Al-Zaidy, S. A., Shell, R., et al. (2017). AVXS-101 phase 1 gene therapy clinical trial in SMA type 1: Event free survival and achievement of developmental milestones [abstract]. Neurology, 88(16 suppl), CT.003. Mendell, J., Al-Zaidy, S. A., Shell, R., et al. (2017). AVXS-101 phase 1 gene therapy clinical trial in SMA type 1: Event free survival and achievement of developmental milestones [abstract]. Neurology, 88(16 suppl), CT.003.
go back to reference Shen, W., Proost, P., Li, B., Gong, W., Le, Y., Sargeant, R., et al. (2000). Activation of the chemotactic peptide receptor FPRL1 in monocytes phosphorylates the chemokine receptor CCR73 and attenuates cell responses to selected chemokines. Biochemical and Biophysical Research Communication, 272(1), 276–283. https://doi.org/10.1006/bbrc.2000.2770.CrossRef Shen, W., Proost, P., Li, B., Gong, W., Le, Y., Sargeant, R., et al. (2000). Activation of the chemotactic peptide receptor FPRL1 in monocytes phosphorylates the chemokine receptor CCR73 and attenuates cell responses to selected chemokines. Biochemical and Biophysical Research Communication, 272(1), 276–283. https://​doi.​org/​10.​1006/​bbrc.​2000.​2770.CrossRef
go back to reference Tonelli, F., Lim, K. G., Loveridge, C., Long, J., Pitson, S. M., Tigyi, G., et al. (2010). FTY720 and (S)-FTY720 vinylphosphonate inhibit sphingosine kinase 1 and promote its proteasomal degradation in human pulmonary artery smooth muscle, breast cancer and androgen-independent prostate cancer cells. Cell Signaling, 22(10), 1536–1542. https://doi.org/10.1016/j.cellsig.2010.05.022.CrossRef Tonelli, F., Lim, K. G., Loveridge, C., Long, J., Pitson, S. M., Tigyi, G., et al. (2010). FTY720 and (S)-FTY720 vinylphosphonate inhibit sphingosine kinase 1 and promote its proteasomal degradation in human pulmonary artery smooth muscle, breast cancer and androgen-independent prostate cancer cells. Cell Signaling, 22(10), 1536–1542. https://​doi.​org/​10.​1016/​j.​cellsig.​2010.​05.​022.CrossRef
go back to reference Van Brocklyn, J. R., & Williams, J. B. (2012). The control of the balance between ceramide and sphingosine-1-phosphate by sphingosine kinase: Oxidative stress and the seesaw of cell survival and death. Comparative Biochemistry and Physiology: Part B, Biochemistry & Molecular Biology, 163(1), 26–36. https://doi.org/10.1016/j.cbpb.2012.05.006.CrossRef Van Brocklyn, J. R., & Williams, J. B. (2012). The control of the balance between ceramide and sphingosine-1-phosphate by sphingosine kinase: Oxidative stress and the seesaw of cell survival and death. Comparative Biochemistry and Physiology: Part B, Biochemistry & Molecular Biology, 163(1), 26–36. https://​doi.​org/​10.​1016/​j.​cbpb.​2012.​05.​006.CrossRef
Metadata
Title
Beneficial Effects of Fingolimod in Alzheimer’s Disease: Molecular Mechanisms and Therapeutic Potential
Authors
Efthalia Angelopoulou
Christina Piperi
Publication date
01-09-2019
Publisher
Springer US
Published in
NeuroMolecular Medicine / Issue 3/2019
Print ISSN: 1535-1084
Electronic ISSN: 1559-1174
DOI
https://doi.org/10.1007/s12017-019-08558-2

Other articles of this Issue 3/2019

NeuroMolecular Medicine 3/2019 Go to the issue