Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2017

Open Access 01-12-2017 | Research

DHCR24 exerts neuroprotection upon inflammation-induced neuronal death

Authors: Henna Martiskainen, Kaisa M. A. Paldanius, Teemu Natunen, Mari Takalo, Mikael Marttinen, Stina Leskelä, Nadine Huber, Petra Mäkinen, Enni Bertling, Hiramani Dhungana, Mikko Huuskonen, Paavo Honkakoski, Pirta Hotulainen, Kirsi Rilla, Jari Koistinaho, Hilkka Soininen, Tarja Malm, Annakaisa Haapasalo, Mikko Hiltunen

Published in: Journal of Neuroinflammation | Issue 1/2017

Login to get access

Abstract

Background

DHCR24, involved in the de novo synthesis of cholesterol and protection of neuronal cells against different stress conditions, has been shown to be selectively downregulated in neurons of the affected brain areas in Alzheimer’s disease.

Methods

Here, we investigated whether the overexpression of DHCR24 protects neurons against inflammation-induced neuronal death using co-cultures of mouse embryonic primary cortical neurons and BV2 microglial cells upon acute neuroinflammation. Moreover, the effects of DHCR24 overexpression on dendritic spine density and morphology in cultured mature mouse hippocampal neurons and on the outcome measures of ischemia-induced brain damage in vivo in mice were assessed.

Results

Overexpression of DHCR24 reduced the loss of neurons under inflammation elicited by LPS and IFN-γ treatment in co-cultures of mouse neurons and BV2 microglial cells but did not affect the production of neuroinflammatory mediators, total cellular cholesterol levels, or the activity of proteins linked with neuroprotective signaling. Conversely, the levels of post-synaptic cell adhesion protein neuroligin-1 were significantly increased upon the overexpression of DHCR24 in basal growth conditions. Augmentation of DHCR24 also increased the total number of dendritic spines and the proportion of mushroom spines in mature mouse hippocampal neurons. In vivo, overexpression of DHCR24 in striatum reduced the lesion size measured by MRI in a mouse model of transient focal ischemia.

Conclusions

These results suggest that the augmentation of DHCR24 levels provides neuroprotection in acute stress conditions, which lead to neuronal loss in vitro and in vivo.
Appendix
Available only for authorised users
Literature
1.
go back to reference Lichtenthaler SF, Haass C, Steiner H. Regulated intramembrane proteolysis—lessons from amyloid precursor protein processing. J Neurochem. 2011;117:779–96.CrossRefPubMed Lichtenthaler SF, Haass C, Steiner H. Regulated intramembrane proteolysis—lessons from amyloid precursor protein processing. J Neurochem. 2011;117:779–96.CrossRefPubMed
2.
go back to reference Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–6.CrossRefPubMed Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–6.CrossRefPubMed
3.
go back to reference Greeve I, Hermans-Borgmeyer I, Brellinger C, Kasper D, Gomez-Isla T, Behl C, et al. The human DIMINUTO/DWARF1 homolog seladin-1 confers resistance to Alzheimer’s disease-associated neurodegeneration and oxidative stress. J Neurosci. 2000;20:7345–52.PubMed Greeve I, Hermans-Borgmeyer I, Brellinger C, Kasper D, Gomez-Isla T, Behl C, et al. The human DIMINUTO/DWARF1 homolog seladin-1 confers resistance to Alzheimer’s disease-associated neurodegeneration and oxidative stress. J Neurosci. 2000;20:7345–52.PubMed
4.
go back to reference Iivonen S, Hiltunen M, Alafuzoff I, Mannermaa A, Kerokoski P, Puoliväli J, et al. Seladin-1 transcription is linked to neuronal degeneration in Alzheimer’s disease. Neuroscience. 2002;113:301–10.CrossRefPubMed Iivonen S, Hiltunen M, Alafuzoff I, Mannermaa A, Kerokoski P, Puoliväli J, et al. Seladin-1 transcription is linked to neuronal degeneration in Alzheimer’s disease. Neuroscience. 2002;113:301–10.CrossRefPubMed
5.
go back to reference Sharpe LJ, Wong J, Garner B, Halliday GM, Brown AJ. Is seladin-1 really a selective Alzheimer’s disease indicator? J Alzheimers Dis. 2012;30:35–9.PubMed Sharpe LJ, Wong J, Garner B, Halliday GM, Brown AJ. Is seladin-1 really a selective Alzheimer’s disease indicator? J Alzheimers Dis. 2012;30:35–9.PubMed
6.
go back to reference Lämsä R, Helisalmi S, Hiltunen M, Herukka S-K, Tapiola T, Pirttilä T, et al. The association study between DHCR24 polymorphisms and Alzheimer’s disease. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:906–10.CrossRefPubMed Lämsä R, Helisalmi S, Hiltunen M, Herukka S-K, Tapiola T, Pirttilä T, et al. The association study between DHCR24 polymorphisms and Alzheimer’s disease. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:906–10.CrossRefPubMed
7.
go back to reference Lu X, Kambe F, Cao X, Kozaki Y, Kaji T, Ishii T, et al. 3beta-Hydroxysteroid-delta24 reductase is a hydrogen peroxide scavenger, protecting cells from oxidative stress-induced apoptosis. Endocrinology. 2008;149:3267–73.CrossRefPubMed Lu X, Kambe F, Cao X, Kozaki Y, Kaji T, Ishii T, et al. 3beta-Hydroxysteroid-delta24 reductase is a hydrogen peroxide scavenger, protecting cells from oxidative stress-induced apoptosis. Endocrinology. 2008;149:3267–73.CrossRefPubMed
8.
go back to reference Waterham HR, Koster J, Romeijn GJ, Hennekam RC, Vreken P, Andersson HC, et al. Mutations in the 3beta-hydroxysterol Delta24-reductase gene cause desmosterolosis, an autosomal recessive disorder of cholesterol biosynthesis. Am J Hum Genet. 2001;69:685–94.CrossRefPubMedPubMedCentral Waterham HR, Koster J, Romeijn GJ, Hennekam RC, Vreken P, Andersson HC, et al. Mutations in the 3beta-hydroxysterol Delta24-reductase gene cause desmosterolosis, an autosomal recessive disorder of cholesterol biosynthesis. Am J Hum Genet. 2001;69:685–94.CrossRefPubMedPubMedCentral
9.
go back to reference Cecchi C, Rosati F, Pensalfini A, Formigli L, Nosi D, Liguri G, et al. Seladin-1/DHCR24 protects neuroblastoma cells against Abeta toxicity by increasing membrane cholesterol content. J Cell Mol Med. 2008;12:1990–2002.CrossRefPubMedPubMedCentral Cecchi C, Rosati F, Pensalfini A, Formigli L, Nosi D, Liguri G, et al. Seladin-1/DHCR24 protects neuroblastoma cells against Abeta toxicity by increasing membrane cholesterol content. J Cell Mol Med. 2008;12:1990–2002.CrossRefPubMedPubMedCentral
10.
go back to reference Crameri A, Biondi E, Kuehnle K, Lütjohann D, Thelen KM, Perga S, et al. The role of seladin-1/DHCR24 in cholesterol biosynthesis, APP processing and Abeta generation in vivo. EMBO J. 2006;25:432–43.CrossRefPubMedPubMedCentral Crameri A, Biondi E, Kuehnle K, Lütjohann D, Thelen KM, Perga S, et al. The role of seladin-1/DHCR24 in cholesterol biosynthesis, APP processing and Abeta generation in vivo. EMBO J. 2006;25:432–43.CrossRefPubMedPubMedCentral
11.
go back to reference Kuehnle K, Crameri A, Kälin RE, Luciani P, Benvenuti S, Peri A, et al. Prosurvival effect of DHCR24/Seladin-1 in acute and chronic responses to oxidative stress. Mol Cell Biol. 2008;28:539–50.CrossRefPubMed Kuehnle K, Crameri A, Kälin RE, Luciani P, Benvenuti S, Peri A, et al. Prosurvival effect of DHCR24/Seladin-1 in acute and chronic responses to oxidative stress. Mol Cell Biol. 2008;28:539–50.CrossRefPubMed
12.
go back to reference Lu X, Li Y, Wang W, Chen S, Liu T, Jia D, et al. 3 β-hydroxysteroid-Δ 24 reductase (DHCR24) protects neuronal cells from apoptotic cell death induced by endoplasmic reticulum (ER) stress. Padmanabhan J, editor. PLoS One. 2014;9:e86753.CrossRefPubMedPubMedCentral Lu X, Li Y, Wang W, Chen S, Liu T, Jia D, et al. 3 β-hydroxysteroid-Δ 24 reductase (DHCR24) protects neuronal cells from apoptotic cell death induced by endoplasmic reticulum (ER) stress. Padmanabhan J, editor. PLoS One. 2014;9:e86753.CrossRefPubMedPubMedCentral
13.
go back to reference Wu C, Miloslavskaya I, Demontis S, Maestro R, Galaktionov K. Regulation of cellular response to oncogenic and oxidative stress by Seladin-1. Nature. 2004;432:640–5.CrossRefPubMed Wu C, Miloslavskaya I, Demontis S, Maestro R, Galaktionov K. Regulation of cellular response to oncogenic and oxidative stress by Seladin-1. Nature. 2004;432:640–5.CrossRefPubMed
14.
15.
go back to reference Abad-Rodriguez J, Ledesma MD, Craessaerts K, Perga S, Medina M, Delacourte A, et al. Neuronal membrane cholesterol loss enhances amyloid peptide generation. J Cell Biol. 2004;167:953–60.CrossRefPubMedPubMedCentral Abad-Rodriguez J, Ledesma MD, Craessaerts K, Perga S, Medina M, Delacourte A, et al. Neuronal membrane cholesterol loss enhances amyloid peptide generation. J Cell Biol. 2004;167:953–60.CrossRefPubMedPubMedCentral
16.
go back to reference Calleros L, Lasa M, Toro MJ, Chiloeches A. Low cell cholesterol levels increase NFkappaB activity through a p38 MAPK-dependent mechanism. Cell Signal. 2006;18:2292–301.CrossRefPubMed Calleros L, Lasa M, Toro MJ, Chiloeches A. Low cell cholesterol levels increase NFkappaB activity through a p38 MAPK-dependent mechanism. Cell Signal. 2006;18:2292–301.CrossRefPubMed
17.
go back to reference Ledesma MD, Abad-Rodriguez J, Galvan C, Biondi E, Navarro P, Delacourte A, et al. Raft disorganization leads to reduced plasmin activity in Alzheimer’s disease brains. EMBO Rep. 2003;4:1190–6.CrossRefPubMedPubMedCentral Ledesma MD, Abad-Rodriguez J, Galvan C, Biondi E, Navarro P, Delacourte A, et al. Raft disorganization leads to reduced plasmin activity in Alzheimer’s disease brains. EMBO Rep. 2003;4:1190–6.CrossRefPubMedPubMedCentral
18.
go back to reference Sarajärvi T, Haapasalo A, Viswanathan J, Mäkinen P, Laitinen M, Soininen H, et al. Down-regulation of seladin-1 increases BACE1 levels and activity through enhanced GGA3 depletion during apoptosis. J Biol Chem. 2009;284:34433–43.CrossRefPubMedPubMedCentral Sarajärvi T, Haapasalo A, Viswanathan J, Mäkinen P, Laitinen M, Soininen H, et al. Down-regulation of seladin-1 increases BACE1 levels and activity through enhanced GGA3 depletion during apoptosis. J Biol Chem. 2009;284:34433–43.CrossRefPubMedPubMedCentral
19.
go back to reference Gresa-Arribas N, Serratosa J, Saura J, Solà C. Inhibition of CCAAT/enhancer binding protein δ expression by chrysin in microglial cells results in anti-inflammatory and neuroprotective effects. J Neurochem. 2010;115:526–36.CrossRefPubMed Gresa-Arribas N, Serratosa J, Saura J, Solà C. Inhibition of CCAAT/enhancer binding protein δ expression by chrysin in microglial cells results in anti-inflammatory and neuroprotective effects. J Neurochem. 2010;115:526–36.CrossRefPubMed
20.
go back to reference Kurkinen KMA, Marttinen M, Turner L, Natunen T, Mäkinen P, Haapalinna F, et al. SEPT8 modulates β-amyloidogenic processing of APP by affecting the sorting and accumulation of BACE1. J Cell Sci. 2016;129:2224–38.CrossRefPubMed Kurkinen KMA, Marttinen M, Turner L, Natunen T, Mäkinen P, Haapalinna F, et al. SEPT8 modulates β-amyloidogenic processing of APP by affecting the sorting and accumulation of BACE1. J Cell Sci. 2016;129:2224–38.CrossRefPubMed
21.
go back to reference Rodriguez A, Ehlenberger DB, Dickstein DL, Hof PR, Wearne SL. Automated three-dimensional detection and shape classification of dendritic spines from fluorescence microscopy images. Sabatini B, editor. PLoS One. 2008;3:e1997.CrossRefPubMedPubMedCentral Rodriguez A, Ehlenberger DB, Dickstein DL, Hof PR, Wearne SL. Automated three-dimensional detection and shape classification of dendritic spines from fluorescence microscopy images. Sabatini B, editor. PLoS One. 2008;3:e1997.CrossRefPubMedPubMedCentral
22.
go back to reference Bertling E, Englund J, Minkeviciene R, Koskinen M, Segerstråle M, Castrén E, et al. Actin Tyrosine-53-Phosphorylation in neuronal maturation and synaptic plasticity. J Neurosci. 2016;36:5299–313.CrossRefPubMed Bertling E, Englund J, Minkeviciene R, Koskinen M, Segerstråle M, Castrén E, et al. Actin Tyrosine-53-Phosphorylation in neuronal maturation and synaptic plasticity. J Neurosci. 2016;36:5299–313.CrossRefPubMed
23.
go back to reference Kanninen K, Heikkinen R, Malm T, Rolova T, Kuhmonen S, Leinonen H, et al. Intrahippocampal injection of a lentiviral vector expressing Nrf2 improves spatial learning in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2009;106:16505–10.CrossRefPubMedPubMedCentral Kanninen K, Heikkinen R, Malm T, Rolova T, Kuhmonen S, Leinonen H, et al. Intrahippocampal injection of a lentiviral vector expressing Nrf2 improves spatial learning in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2009;106:16505–10.CrossRefPubMedPubMedCentral
24.
go back to reference Dénes A, Humphreys N, Lane TE, Grencis R, Rothwell N. Chronic systemic infection exacerbates ischemic brain damage via a CCL5 (regulated on activation, normal T-cell expressed and secreted)-mediated proinflammatory response in mice. J Neurosci. 2010;30:10086–95.CrossRefPubMedPubMedCentral Dénes A, Humphreys N, Lane TE, Grencis R, Rothwell N. Chronic systemic infection exacerbates ischemic brain damage via a CCL5 (regulated on activation, normal T-cell expressed and secreted)-mediated proinflammatory response in mice. J Neurosci. 2010;30:10086–95.CrossRefPubMedPubMedCentral
25.
go back to reference Lemarchant S, Dunghana H, Pomeshchik Y, Leinonen H, Kolosowska N, Korhonen P, et al. Anti-inflammatory effects of ADAMTS-4 in a mouse model of ischemic stroke. Glia. 2016;64:1492–507.CrossRefPubMed Lemarchant S, Dunghana H, Pomeshchik Y, Leinonen H, Kolosowska N, Korhonen P, et al. Anti-inflammatory effects of ADAMTS-4 in a mouse model of ischemic stroke. Glia. 2016;64:1492–507.CrossRefPubMed
26.
go back to reference Shuaib A, Xu Wang C, Yang T, Noor R. Effects of nonpeptide V(1) vasopressin receptor antagonist SR-49059 on infarction volume and recovery of function in a focal embolic stroke model. Stroke. 2002;33:3033–7.CrossRefPubMed Shuaib A, Xu Wang C, Yang T, Noor R. Effects of nonpeptide V(1) vasopressin receptor antagonist SR-49059 on infarction volume and recovery of function in a focal embolic stroke model. Stroke. 2002;33:3033–7.CrossRefPubMed
27.
go back to reference Brunet A, Datta SR, Greenberg ME. Transcription-dependent and -independent control of neuronal survival by the PI3K-Akt signaling pathway. Curr Opin Neurobiol. 2001;11:297–305.CrossRefPubMed Brunet A, Datta SR, Greenberg ME. Transcription-dependent and -independent control of neuronal survival by the PI3K-Akt signaling pathway. Curr Opin Neurobiol. 2001;11:297–305.CrossRefPubMed
28.
go back to reference Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 2011;75:50–83.CrossRefPubMedPubMedCentral Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 2011;75:50–83.CrossRefPubMedPubMedCentral
29.
go back to reference Mayr B, Montminy M. Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat Rev Mol Cell Biol. 2001;2:599–609.CrossRefPubMed Mayr B, Montminy M. Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat Rev Mol Cell Biol. 2001;2:599–609.CrossRefPubMed
30.
go back to reference Liu A, Zhou Z, Dang R, Zhu Y, Qi J, He G, et al. Neuroligin 1 regulates spines and synaptic plasticity via LIMK1/cofilin-mediated actin reorganization. J Cell Biol. 2016;212:449–63.CrossRefPubMedPubMedCentral Liu A, Zhou Z, Dang R, Zhu Y, Qi J, He G, et al. Neuroligin 1 regulates spines and synaptic plasticity via LIMK1/cofilin-mediated actin reorganization. J Cell Biol. 2016;212:449–63.CrossRefPubMedPubMedCentral
31.
go back to reference Segatto M, Leboffe L, Trapani L, Pallottini V. Cholesterol homeostasis failure in the brain: implications for synaptic dysfunction and cognitive decline. Curr Med Chem. 2014;21:2788–802.CrossRefPubMed Segatto M, Leboffe L, Trapani L, Pallottini V. Cholesterol homeostasis failure in the brain: implications for synaptic dysfunction and cognitive decline. Curr Med Chem. 2014;21:2788–802.CrossRefPubMed
32.
go back to reference Maulik M, Westaway D, Jhamandas JH, Kar S. Role of cholesterol in APP metabolism and its significance in Alzheimer’s disease pathogenesis. Mol Neurobiol. 2013;47:37–63.CrossRefPubMed Maulik M, Westaway D, Jhamandas JH, Kar S. Role of cholesterol in APP metabolism and its significance in Alzheimer’s disease pathogenesis. Mol Neurobiol. 2013;47:37–63.CrossRefPubMed
33.
go back to reference Tesco G, Koh YH, Tanzi RE. Caspase activation increases beta-amyloid generation independently of caspase cleavage of the beta-amyloid precursor protein (APP). J Biol Chem. 2003;278:46074–80.CrossRefPubMed Tesco G, Koh YH, Tanzi RE. Caspase activation increases beta-amyloid generation independently of caspase cleavage of the beta-amyloid precursor protein (APP). J Biol Chem. 2003;278:46074–80.CrossRefPubMed
34.
go back to reference Hering H, Lin C-C, Sheng M. Lipid rafts in the maintenance of synapses, dendritic spines, and surface AMPA receptor stability. J Neurosci. 2003;23:3262–71.PubMed Hering H, Lin C-C, Sheng M. Lipid rafts in the maintenance of synapses, dendritic spines, and surface AMPA receptor stability. J Neurosci. 2003;23:3262–71.PubMed
35.
go back to reference Park H, Poo M. Neurotrophin regulation of neural circuit development and function. Nat Rev Neurosci. 2013;14:7–23.CrossRefPubMed Park H, Poo M. Neurotrophin regulation of neural circuit development and function. Nat Rev Neurosci. 2013;14:7–23.CrossRefPubMed
36.
go back to reference Venugopal R, Jaiswal AK. Nrf1 and Nrf2 positively and c-Fos and Fra1 negatively regulate the human antioxidant response element-mediated expression of NAD(P)H:quinone oxidoreductase1 gene. Proc Natl Acad Sci U S A. 1996;93:14960–5.CrossRefPubMedPubMedCentral Venugopal R, Jaiswal AK. Nrf1 and Nrf2 positively and c-Fos and Fra1 negatively regulate the human antioxidant response element-mediated expression of NAD(P)H:quinone oxidoreductase1 gene. Proc Natl Acad Sci U S A. 1996;93:14960–5.CrossRefPubMedPubMedCentral
37.
go back to reference Minelli A, Conte C, Grottelli S, Bellezza I, Emiliani C, Bolaños JP. Cyclo(his-pro) up-regulates heme oxygenase 1 via activation of Nrf2-ARE signalling. J Neurochem. 2009;111:956–66.CrossRefPubMed Minelli A, Conte C, Grottelli S, Bellezza I, Emiliani C, Bolaños JP. Cyclo(his-pro) up-regulates heme oxygenase 1 via activation of Nrf2-ARE signalling. J Neurochem. 2009;111:956–66.CrossRefPubMed
38.
go back to reference Ko J, Zhang C, Arac D, Boucard AA, Brunger AT, Südhof TC. Neuroligin-1 performs neurexin-dependent and neurexin-independent functions in synapse validation. EMBO J. 2009;28:3244–55.CrossRefPubMedPubMedCentral Ko J, Zhang C, Arac D, Boucard AA, Brunger AT, Südhof TC. Neuroligin-1 performs neurexin-dependent and neurexin-independent functions in synapse validation. EMBO J. 2009;28:3244–55.CrossRefPubMedPubMedCentral
39.
go back to reference Kwon H-B, Kozorovitskiy Y, Oh W-J, Peixoto RT, Akhtar N, Saulnier JL, et al. Neuroligin-1-dependent competition regulates cortical synaptogenesis and synapse number. Nat Neurosci. 2012;15:1667–74.CrossRefPubMedPubMedCentral Kwon H-B, Kozorovitskiy Y, Oh W-J, Peixoto RT, Akhtar N, Saulnier JL, et al. Neuroligin-1-dependent competition regulates cortical synaptogenesis and synapse number. Nat Neurosci. 2012;15:1667–74.CrossRefPubMedPubMedCentral
41.
go back to reference Martin MG, Ahmed T, Korovaichuk A, Venero C, Menchón SA, Salas I, et al. Constitutive hippocampal cholesterol loss underlies poor cognition in old rodents. EMBO Mol Med. 2014;6:902–17.CrossRefPubMedPubMedCentral Martin MG, Ahmed T, Korovaichuk A, Venero C, Menchón SA, Salas I, et al. Constitutive hippocampal cholesterol loss underlies poor cognition in old rodents. EMBO Mol Med. 2014;6:902–17.CrossRefPubMedPubMedCentral
42.
go back to reference Korinek M, Vyklicky V, Borovska J, Lichnerova K, Kaniakova M, Krausova B, et al. Cholesterol modulates open probability and desensitization of NMDA receptors. J Physiol. 2015;593:2279–93.CrossRefPubMedPubMedCentral Korinek M, Vyklicky V, Borovska J, Lichnerova K, Kaniakova M, Krausova B, et al. Cholesterol modulates open probability and desensitization of NMDA receptors. J Physiol. 2015;593:2279–93.CrossRefPubMedPubMedCentral
43.
go back to reference Parsons MP, Raymond LA. Extrasynaptic NMDA receptor involvement in central nervous system disorders. Neuron. 2014;82:279–93.CrossRefPubMed Parsons MP, Raymond LA. Extrasynaptic NMDA receptor involvement in central nervous system disorders. Neuron. 2014;82:279–93.CrossRefPubMed
44.
go back to reference Kern A, Roempp B, Prager K, Walter J, Behl C. Down-regulation of endogenous amyloid precursor protein processing due to cellular aging. J Biol Chem. 2006;281:2405–13.CrossRefPubMed Kern A, Roempp B, Prager K, Walter J, Behl C. Down-regulation of endogenous amyloid precursor protein processing due to cellular aging. J Biol Chem. 2006;281:2405–13.CrossRefPubMed
45.
go back to reference Molander-Melin M, Blennow K, Bogdanovic N, Dellheden B, Månsson J-E, Fredman P. Structural membrane alterations in Alzheimer brains found to be associated with regional disease development; increased density of gangliosides GM1 and GM2 and loss of cholesterol in detergent-resistant membrane domains. J Neurochem. 2005;92:171–82.CrossRefPubMed Molander-Melin M, Blennow K, Bogdanovic N, Dellheden B, Månsson J-E, Fredman P. Structural membrane alterations in Alzheimer brains found to be associated with regional disease development; increased density of gangliosides GM1 and GM2 and loss of cholesterol in detergent-resistant membrane domains. J Neurochem. 2005;92:171–82.CrossRefPubMed
46.
go back to reference Roher AE, Weiss N, Kokjohn TA, Kuo Y-M, Kalback W, Anthony J, et al. Increased a beta peptides and reduced cholesterol and myelin proteins characterize white matter degeneration in Alzheimer’s disease. Biochemistry. 2002;41:11080–90.CrossRefPubMed Roher AE, Weiss N, Kokjohn TA, Kuo Y-M, Kalback W, Anthony J, et al. Increased a beta peptides and reduced cholesterol and myelin proteins characterize white matter degeneration in Alzheimer’s disease. Biochemistry. 2002;41:11080–90.CrossRefPubMed
47.
go back to reference Thelen KM, Falkai P, Bayer TA, Lütjohann D. Cholesterol synthesis rate in human hippocampus declines with aging. Neurosci Lett. 2006;403:15–9.CrossRefPubMed Thelen KM, Falkai P, Bayer TA, Lütjohann D. Cholesterol synthesis rate in human hippocampus declines with aging. Neurosci Lett. 2006;403:15–9.CrossRefPubMed
48.
go back to reference Ichtchenko K, Hata Y, Nguyen T, Ullrich B, Missler M, Moomaw C, et al. Neuroligin 1: a splice site-specific ligand for beta-neurexins. Cell. 1995;81:435–43.CrossRefPubMed Ichtchenko K, Hata Y, Nguyen T, Ullrich B, Missler M, Moomaw C, et al. Neuroligin 1: a splice site-specific ligand for beta-neurexins. Cell. 1995;81:435–43.CrossRefPubMed
49.
go back to reference Hernández-Jiménez M, Martínez-López D, Gabandé-Rodríguez E, Martín-Segura A, Lizasoain I, Ledesma MD, et al. Seladin-1/DHCR24 is Neuroprotective by associating EAAT2 glutamate transporter to lipid rafts in experimental stroke. Stroke. 2016;47:206–13.CrossRefPubMed Hernández-Jiménez M, Martínez-López D, Gabandé-Rodríguez E, Martín-Segura A, Lizasoain I, Ledesma MD, et al. Seladin-1/DHCR24 is Neuroprotective by associating EAAT2 glutamate transporter to lipid rafts in experimental stroke. Stroke. 2016;47:206–13.CrossRefPubMed
Metadata
Title
DHCR24 exerts neuroprotection upon inflammation-induced neuronal death
Authors
Henna Martiskainen
Kaisa M. A. Paldanius
Teemu Natunen
Mari Takalo
Mikael Marttinen
Stina Leskelä
Nadine Huber
Petra Mäkinen
Enni Bertling
Hiramani Dhungana
Mikko Huuskonen
Paavo Honkakoski
Pirta Hotulainen
Kirsi Rilla
Jari Koistinaho
Hilkka Soininen
Tarja Malm
Annakaisa Haapasalo
Mikko Hiltunen
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2017
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-017-0991-6

Other articles of this Issue 1/2017

Journal of Neuroinflammation 1/2017 Go to the issue