Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Int J Mol Sci ; 24(3)2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36769295

ABSTRACT

Gemfibrozil is a drug that has been used for over 40 years to lower triglycerides in blood. As a ligand for peroxisome proliferative-activated receptor-alpha (PPARα), which is expressed in many tissues, it induces the transcription of numerous genes for carbohydrate and lipid-metabolism. However, nothing is known about how intracellular lipid-homeostasis and, in particular, triglycerides are affected. As triglycerides are stored in lipid-droplets, which are known to be associated with many diseases, such as Alzheimer's disease, cancer, fatty liver disease and type-2 diabetes, treatment with gemfibrozil could adversely affect these diseases. To address the question whether gemfibrozil also affects intracellular lipid-levels, SH-SY5Y, HEK and Calu-3 cells, representing three different metabolically active organs (brain, lung and kidney), were incubated with gemfibrozil and subsequently analyzed semi-quantitatively by mass-spectrometry. Importantly, all cells showed a strong increase in intracellular triglycerides (SH-SY5Y: 170.3%; HEK: 272.1%; Calu-3: 448.1%), suggesting that the decreased triglyceride-levels might be due to an enhanced cellular uptake. Besides the common intracellular triglyceride increase, a cell-line specific alteration in acylcarnitines are found, suggesting that especially in neuronal cell lines gemfibrozil increases the transport of fatty acids to mitochondria and therefore increases the turnover of fatty acids for the benefit of additional energy supply, which could be important in diseases, such as Alzheimer's disease.


Subject(s)
Alzheimer Disease , Neuroblastoma , Humans , Gemfibrozil/pharmacology , Alzheimer Disease/drug therapy , Neuroblastoma/drug therapy , Triglycerides/metabolism , Fatty Acids , Hypolipidemic Agents/pharmacology , Hypolipidemic Agents/therapeutic use
2.
Int J Mol Sci ; 23(4)2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35216410

ABSTRACT

Alzheimer's disease (AD) is characterized by an increased plaque burden and tangle accumulation in the brain accompanied by extensive lipid alterations. Methylxanthines (MTXs) are alkaloids frequently consumed by dietary intake known to interfere with the molecular mechanisms leading to AD. Besides the fact that MTX consumption is associated with changes in triglycerides and cholesterol in serum and liver, little is known about the effect of MTXs on other lipid classes, which raises the question of whether MTX can alter lipids in a way that may be relevant in AD. Here we have analyzed naturally occurring MTXs caffeine, theobromine, theophylline, and the synthetic MTXs pentoxifylline and propentofylline also used as drugs in different neuroblastoma cell lines. Our results show that lipid alterations are not limited to triglycerides and cholesterol in the liver and serum, but also include changes in sphingomyelins, ceramides, phosphatidylcholine, and plasmalogens in neuroblastoma cells. These changes comprise alterations known to be beneficial, but also adverse effects regarding AD were observed. Our results give an additional perspective of the complex link between MTX and AD, and suggest combining MTX with a lipid-altering diet compensating the adverse effects of MTX rather than using MTX alone to prevent or treat AD.


Subject(s)
Alzheimer Disease/metabolism , Lipids/physiology , Neuroblastoma/metabolism , Neurodegenerative Diseases/metabolism , Xanthines/pharmacology , Caffeine/pharmacology , Cell Line, Tumor , Cholesterol/metabolism , Humans , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Pentoxifylline/pharmacology , Theobromine/pharmacology , Theophylline/pharmacology , Triglycerides/metabolism
3.
Int J Mol Sci ; 23(24)2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36555176

ABSTRACT

Administration of systemic retinoids such as acitretin has not been approved yet for pediatric patients. An adverse event of retinoid-therapy that occurs with lower prevalence in children than in adults is hyperlipidemia. This might be based on the lack of comorbidities in young patients, but must not be neglected. Especially for the development of the human brain up to young adulthood, dysbalance of lipids might be deleterious. Here, we provide for the first time an in-depth analysis of the influence of subchronic acitretin-administration on lipid composition of brain parenchyma of young wild type mice. For comparison and to evaluate the systemic effect of the treatment, liver lipids were analogously investigated. As expected, triglycerides increased in liver as well as in brain and a non-significant increase in cholesterol was observed. However, specifically brain showed an increase in lyso-phosphatidylcholine and carnitine as well as in sphingomyelin. Group analysis of lipid classes revealed no statistical effects, while single species were tissue-dependently changed: effects in brain were in general more subtly as compared to those in liver regarding the mere number of changed lipid species. Thus, while the overall impact of acitretin seems comparably small regarding brain, the change in individual species and their role in brain development and maturation has to be considered.


Subject(s)
Acitretin , Hyperlipidemias , Adult , Humans , Child , Adolescent , Animals , Mice , Young Adult , Acitretin/pharmacology , Acitretin/therapeutic use , Lipidomics , Hyperlipidemias/chemically induced , Cholesterol , Brain
4.
Cell Mol Life Sci ; 76(5): 1005-1025, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30599067

ABSTRACT

BACKGROUND: The ADAM10-mediated cleavage of transmembrane proteins regulates cellular processes such as proliferation or migration. Substrate cleavage by ADAM10 has also been implicated in pathological situations such as cancer or Morbus Alzheimer. Therefore, identifying endogenous molecules, which modulate the amount and consequently the activity of ADAM10, might contribute to a deeper understanding of the enzyme's role in both, physiology and pathology. METHOD: To elucidate the underlying cellular mechanism of the TBX2-mediated repression of ADAM10 gene expression, we performed overexpression, RNAi-mediated knockdown and pharmacological inhibition studies in the human neuroblastoma cell line SH-SY5Y. Expression analysis was conducted by e.g. real-time RT-PCR or western blot techniques. To identify the binding region of TBX2 within the ADAM10 promoter, we used luciferase reporter assay on deletion constructs and EMSA/WEMSA experiments. In addition, we analyzed a TBX2 loss-of-function Drosophila model regarding the expression of ADAM10 orthologs by qPCR. Furthermore, we quantified the mRNA level of TBX2 in post-mortem brain tissue of AD patients. RESULTS: Here, we report TBX2 as a transcriptional repressor of ADAM10 gene expression: both, the DNA-binding domain and the repression domain of TBX2 were necessary to effect transcriptional repression of ADAM10 in neuronal SH-SY5Y cells. This regulatory mechanism required HDAC1 as a co-factor of TBX2. Transcriptional repression was mediated by two functional TBX2 binding sites within the core promoter sequence (- 315 to - 286 bp). Analysis of a TBX2 loss-of-function Drosophila model revealed that kuzbanian and kuzbanian-like, orthologs of ADAM10, were derepressed compared to wild type. Vice versa, analysis of cortical brain samples of AD-patients, which showed reduced ADAM10 mRNA levels, revealed a 2.5-fold elevation of TBX2, while TBX3 and TBX21 levels were not affected. CONCLUSION: Our results characterize TBX2 as a repressor of ADAM10 gene expression and suggest that this regulatory interaction is conserved across tissues and species.


Subject(s)
ADAM10 Protein/genetics , Alzheimer Disease/etiology , Gene Expression Regulation , T-Box Domain Proteins/physiology , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Binding Sites , Brain/metabolism , Cells, Cultured , Disintegrins/genetics , Drosophila , Drosophila Proteins/genetics , Histone Deacetylase 1/physiology , Humans , Metalloendopeptidases/genetics , Mice , Mice, Inbred C57BL , Neurons/metabolism , Promoter Regions, Genetic , T-Box Domain Proteins/chemistry , Transcription, Genetic
5.
Int J Mol Sci ; 21(23)2020 Nov 27.
Article in English | MEDLINE | ID: mdl-33260941

ABSTRACT

Methylxanthines are a group of substances derived from the purine base xanthine with a methyl group at the nitrogen on position 3 and different residues at the nitrogen on position 1 and 7. They are widely consumed in nutrition and used as pharmaceuticals. Here we investigate the transcriptional regulation of 83 genes linked to Alzheimer's disease in the presence of five methylxanthines, including the most prominent naturally occurring methylxanthines-caffeine, theophylline and theobromine-and the synthetic methylxanthines pentoxifylline and propentofylline. Methylxanthine-regulated genes were found in pathways involved in processes including oxidative stress, lipid homeostasis, signal transduction, transcriptional regulation, as well as pathways involved in neuronal function. Interestingly, multivariate analysis revealed different or inverse effects on gene regulation for caffeine compared to the other methylxanthines, which was further substantiated by multiple comparison analysis, pointing out a distinct role for caffeine in gene regulation. Our results not only underline the beneficial effects of methylxanthines in the regulation of genes in neuroblastoma wild-type cells linked to neurodegenerative diseases in general, but also demonstrate that individual methylxanthines like caffeine mediate unique or inverse expression patterns. This suggests that the replacement of single methylxanthines by others could result in unexpected effects, which could not be anticipated by the comparison to other substances in this substance class.


Subject(s)
Alzheimer Disease/genetics , Caffeine/pharmacology , Gene Expression Regulation/drug effects , Neuroblastoma/genetics , Xanthines/pharmacology , Cell Line, Tumor , Genes, Essential , Humans , Pentoxifylline/pharmacology , Principal Component Analysis , Theobromine/pharmacology , Theophylline/pharmacology , Transcription, Genetic/drug effects , Xanthines/chemistry
6.
J Lipid Res ; 58(11): 2083-2101, 2017 11.
Article in English | MEDLINE | ID: mdl-28528321

ABSTRACT

In the last decade, it has become obvious that Alzheimer's disease (AD) is closely linked to changes in lipids or lipid metabolism. One of the main pathological hallmarks of AD is amyloid-ß (Aß) deposition. Aß is derived from sequential proteolytic processing of the amyloid precursor protein (APP). Interestingly, both, the APP and all APP secretases are transmembrane proteins that cleave APP close to and in the lipid bilayer. Moreover, apoE4 has been identified as the most prevalent genetic risk factor for AD. ApoE is the main lipoprotein in the brain, which has an abundant role in the transport of lipids and brain lipid metabolism. Several lipidomic approaches revealed changes in the lipid levels of cerebrospinal fluid or in post mortem AD brains. Here, we review the impact of apoE and lipids in AD, focusing on the major brain lipid classes, sphingomyelin, plasmalogens, gangliosides, sulfatides, DHA, and EPA, as well as on lipid signaling molecules, like ceramide and sphingosine-1-phosphate. As nutritional approaches showed limited beneficial effects in clinical studies, the opportunities of combining different supplements in multi-nutritional approaches are discussed and summarized.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Apolipoproteins E/metabolism , Fatty Acids, Omega-3/metabolism , Food , Alzheimer Disease/diet therapy , Animals , Humans
7.
Int J Mol Sci ; 18(12)2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29257109

ABSTRACT

Alzheimer's disease (AD) is characterized by extracellular plaques in the brain, mainly consisting of amyloid-ß (Aß), as derived from sequential cleavage of the amyloid precursor protein. Epidemiological studies suggest a tight link between hypovitaminosis of the secosteroid vitamin D and AD. Besides decreased vitamin D level in AD patients, an effect of vitamin D on Aß-homeostasis is discussed. However, the exact underlying mechanisms remain to be elucidated and nothing is known about the potential effect of vitamin D analogues. Here we systematically investigate the effect of vitamin D and therapeutically used analogues (maxacalcitol, calcipotriol, alfacalcidol, paricalcitol, doxercalciferol) on AD-relevant mechanisms. D2 and D3 analogues decreased Aß-production and increased Aß-degradation in neuroblastoma cells or vitamin D deficient mouse brains. Effects were mediated by affecting the Aß-producing enzymes BACE1 and γ-secretase. A reduced secretase activity was accompanied by a decreased BACE1 protein level and nicastrin expression, an essential component of the γ-secretase. Vitamin D and analogues decreased ß-secretase activity, not only in mouse brains with mild vitamin D hypovitaminosis, but also in non-deficient mouse brains. Our results further strengthen the link between AD and vitamin D, suggesting that supplementation of vitamin D or vitamin D analogues might have beneficial effects in AD prevention.


Subject(s)
Amyloid beta-Peptides/metabolism , Plaque, Amyloid/drug therapy , Proteolysis , Vitamin D/therapeutic use , Vitamins/therapeutic use , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , Vitamin D/administration & dosage , Vitamin D/pharmacology , Vitamins/administration & dosage , Vitamins/pharmacology
8.
Biochem Cell Biol ; 94(6): 534-542, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27813426

ABSTRACT

Omega-3 polyunsaturated fatty acids (PUFAs) have been proposed to be highly beneficial in Alzheimer's disease (AD). AD pathology is closely linked to an overproduction and accumulation of amyloid-ß (Aß) peptides as extracellular senile plaques in the brain. Total Aß levels are not only dependent on its production by proteolytic processing of the amyloid precursor protein (APP), but also on Aß-clearance mechanisms, including Aß-degrading enzymes. Here we show that the omega-3 PUFAs eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) increase Aß-degradation by affecting insulin-degrading enzyme (IDE), the major Aß-degrading enzyme secreted into the extracellular space of neuronal and microglial cells. The identification of the molecular mechanisms revealed that EPA directly increases IDE enzyme activity and elevates gene expression of IDE. DHA also directly stimulates IDE enzyme activity and affects IDE sorting by increasing exosome release of IDE, resulting in enhanced Aß-degradation in the extracellular milieu. Apart from the known positive effect of DHA in reducing Aß production, EPA and DHA might ameliorate AD pathology by increasing Aß turnover.


Subject(s)
Amyloid beta-Peptides/metabolism , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Insulysin/genetics , Neuroblastoma/metabolism , Animals , Blotting, Western , Cell Survival/drug effects , Insulysin/metabolism , Mice , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
BMC Neurosci ; 17(1): 44, 2016 07 04.
Article in English | MEDLINE | ID: mdl-27377996

ABSTRACT

BACKGROUND: Murine models of Alzheimer's disease (AD) are mainly based on overexpression of pathologic amyloid precursor protein and/or presenilins. Those genes resemble underlying cause of early onset type of AD while about 99 % of all human cases are to be characterized as sporadic, late onset. Appropriate animal models for this type of AD are still missing. We here investigated, if transnasal delivery of A-beta 42 peptides might serve to mimic pathological effects in mice. RESULTS: A-beta 42 peptides, used for the behavioral study, showed the expected dose-dependent toxicity in neur oblastoma cell line SH-SY5Y and were able to form higher molecular weight species in vitro. Upon delivery into nostrils of wild type mice, protein bands that might represent aggregation products of the exogenously applied human A-beta 42 were only observed in total brain homogenates from mice pre-treated with mannitol. By using TAMRA-labeled A-beta 42 peptides we demonstrated, that transport throughout the brain was achieved already 1 h after administration. FVB/N mice treated with A-beta 42 for 3 days were significantly impaired in the cue-retention condition of the fear conditioning task as compared to controls whereas A-beta-treated C57B6/J mice were impaired in the context condition. In the Morris water maze test, these mice also displayed a delayed learning performance, indicated by significantly longer time to find the platform. Those deficits were also seen for memory performance in the probe trial as measured by number of crossings of the former platform position and time spent in the goal quadrant. CONCLUSIONS: Existing AD mouse models are of genetic origin and need prolonged housing time before onset of pathology. Our short-term treatment induced learning and memory deficits via exogenous application of A-beta peptides comparable to those observed for the transgenic animals. With the transnasal A-beta 42 treatment we present an approach to investigate purely A-beta related changes suitable as a model for symptoms of Alzheimer's dementia (AD). Resulting behavioral deficits were indicative for familial type of Alzheimer's disease as well as for the late onset variant.


Subject(s)
Amyloid beta-Peptides , Disease Models, Animal , Learning Disabilities , Memory Disorders , Peptide Fragments , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Administration, Intranasal , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Learning Disabilities/metabolism , Learning Disabilities/pathology , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Species Specificity
10.
Ann Neurol ; 78(1): 88-103, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25893674

ABSTRACT

OBJECTIVE: Alzheimer's disease (AD)-associated dementia is due to tissue damage caused by amyloid ß (Aß) deposition within the brain and by accompanying neuroinflammation. The nicotinamide adenine dinucleotide (NAD) glycohydrolase CD38, which is expressed by neurons, astrocytes, and microglial cells, regulates inflammatory and repair processes in the brain and other tissues by degrading NAD and repressing the activity of other NAD-consuming enzymes and by producing NAD-derived metabolites that regulate calcium signaling and migration of inflammatory cells. Given the role of CD38 in neuroinflammation and repair, we examined the effect of CD38 deletion on AD pathology. METHODS: We crossed APPswePS1ΔE9 (APP.PS) mice with Cd38(-) (/) (-) mice to generate AD-prone CD38-deficient animals (APP.PS.Cd38(-) (/) (-) ) and examined AD-related phenotypes in both groups. RESULTS: APP.PS.Cd38(-) (/) (-) mice exhibited significant reductions in Aß plaque load and soluble Aß levels compared to APP.PS mice, and this correlated with improved spatial learning. Although CD38 deficiency resulted in decreased microglia/macrophage (MM) accumulation, the transcription profile of the Cd38(-) (/) (-) and Cd38(+/) (+) MM was similar, suggesting that the decreased Aß burden in APP.PS.Cd38(-) (/) (-) mice was not due to alterations in MM activation/function. Instead, APP.PS.Cd38(-) (/) (-) neuronal cultures secreted less Aß and this reduction was mimicked when APP.PS neuronal cultures were treated with inhibitors that blocked CD38 enzyme activity or the signaling pathways controlled by CD38-derived metabolites. Furthermore, ß- and γ-secretase activity was decreased in APP.PS.Cd38(-) (/) (-) mice, which correlated with decreased Aß production. INTERPRETATION: CD38 regulates AD pathology in the APP.PS model of AD, suggesting that CD38 may be a novel target for AD treatment.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Behavior, Animal , Brain/pathology , Membrane Glycoproteins/genetics , Plaque, Amyloid/pathology , RNA, Messenger/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Cell Movement , Cells, Cultured , Disease Models, Animal , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/metabolism , Spatial Learning , Transcriptome
11.
Neurodegener Dis ; 16(1-2): 44-54, 2016.
Article in English | MEDLINE | ID: mdl-26642316

ABSTRACT

One of the main characteristics of Alzheimer's disease (AD) is the ß-amyloid peptide (Aß) generated by ß- and γ-secretase processing of the amyloid precursor protein (APP). Previously it has been demonstrated that polyunsaturated fatty acids (PUFAs), especially docosahexaenoic acid (DHA), are associated with a reduced risk of AD caused by decreased Aß production. However, in epidemiological studies and nutritional approaches, the outcomes of DHA-dependent treatment were partially controversial. PUFAs are very susceptible to reactive oxygen species and lipid peroxidation, which are increased during disease pathology. In line with published results, lipid peroxidation was elevated in human postmortem AD brains; especially 4-hydroxy-nonenal (HNE) was increased. To investigate whether lipid peroxidation is only a consequence or might also influence the processes leading to AD, we analyzed 7 different oxidized lipid species including 5 oxidized DHA derivatives and the lipid peroxidation products of ω-3 and ω-6 PUFAs, HNE and 4-hydroxy-hexenal, in human neuroblastoma cells and mouse mixed cortical neurons. In the presence of oxidized lipids Aß and soluble ß-secreted APP levels were elevated, whereas soluble α-secreted APP was decreased, suggesting a shift from the nonamyloidogenic to the amyloidogenic pathway of APP processing. Furthermore, ß- and γ-secretase activity was increased by oxidized lipids via increased gene expression and additionally by a direct effect on ß-secretase activity. Importantly, only 1% oxidized DHA was sufficient to revert the protective effect of DHA and to significantly increase Aß production. Therefore, our results emphasize the need to prevent DHA from oxidation in nutritional approaches and might help explain the divergent results of clinical DHA studies.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Docosahexaenoic Acids/analogs & derivatives , Docosahexaenoic Acids/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lipid Peroxidation , Male , Mass Spectrometry , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Real-Time Polymerase Chain Reaction , Tissue Banks
12.
Int J Mol Sci ; 17(11)2016 Oct 26.
Article in English | MEDLINE | ID: mdl-27792188

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia in the elderly population, currently affecting 46 million people worldwide. Histopathologically, the disease is characterized by the occurrence of extracellular amyloid plaques composed of aggregated amyloid-ß (Aß) peptides and intracellular neurofibrillary tangles containing the microtubule-associated protein tau. Aß peptides are derived from the sequential processing of the amyloid precursor protein (APP) by enzymes called secretases, which are strongly influenced by the lipid environment. Several vitamins have been reported to be reduced in the plasma/serum of AD-affected individuals indicating they have an impact on AD pathogenesis. In this review we focus on vitamin E and the other lipophilic vitamins A, D, and K, and summarize the current knowledge about their status in AD patients, their impact on cognitive functions and AD risk, as well as their influence on the molecular mechanisms of AD. The vitamins might affect the generation and clearance of Aß both by direct effects and indirectly by altering the cellular lipid homeostasis. Additionally, vitamins A, D, E, and K are reported to influence further mechanisms discussed to be involved in AD pathogenesis, e.g., Aß-aggregation, Aß-induced neurotoxicity, oxidative stress, and inflammatory processes, as summarized in this article.


Subject(s)
Alzheimer Disease/metabolism , Vitamin A/metabolism , Vitamin D/metabolism , Vitamin E/metabolism , Vitamin K/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Humans , Lipid Metabolism
13.
Int J Mol Sci ; 17(11)2016 Oct 29.
Article in English | MEDLINE | ID: mdl-27801864

ABSTRACT

One of the characteristics of Alzheimer´s disease (AD) is an increased amyloid load and an enhanced level of reactive oxidative species (ROS). Vitamin E has known beneficial neuroprotective effects, and previously, some studies suggested that vitamin E is associated with a reduced risk of AD due to its antioxidative properties. However, epidemiological studies and nutritional approaches of vitamin E treatment are controversial. Here, we investigate the effect of α-tocotrienol, which belongs to the group of vitamin E, on AD-relevant processes in neuronal cell lines. In line with the literature, α-tocotrienol reduced the ROS level in SH-SY5Y cells. In the presence of tocotrienols, cholesterol and cholesterol esters, which have been shown to be risk factors in AD, were decreased. Besides the unambiguous positive effects of tocotrienol, amyloid-ß (Aß) levels were increased accompanied by an increase in the activity of enzymes responsible for Aß production. Proteins and gene expression of the secretases and their components remained unchanged, whereas tocotrienol accelerates enzyme activity in cell-free assays. Besides enhanced Aß production, tocotrienols inhibited Aß degradation in neuro 2a (N2a)-cells. Our results might help to understand the controversial findings of vitamin E studies and demonstrate that besides the known positive neuroprotective properties, tocotrienols also have negative characteristics with respect to AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Neuroblastoma/drug therapy , Oxidative Stress/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/biosynthesis , Antioxidants/administration & dosage , Cell Line , Cholesterol/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Reactive Oxygen Species/metabolism , Tocotrienols/administration & dosage , Vitamin E/administration & dosage
14.
J Biol Chem ; 289(24): 16761-72, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24808180

ABSTRACT

Progressive accumulation of the amyloid ß protein in extracellular plaques is a neuropathological hallmark of Alzheimer disease. Amyloid ß is generated during sequential cleavage of the amyloid precursor protein (APP) by ß- and γ-secretases. In addition to the proteolytic processing by secretases, APP is also metabolized by lysosomal proteases. Here, we show that accumulation of intracellular sphingosine-1-phosphate (S1P) impairs the metabolism of APP. Cells lacking functional S1P-lyase, which degrades intracellular S1P, strongly accumulate full-length APP and its potentially amyloidogenic C-terminal fragments (CTFs) as compared with cells expressing the functional enzyme. By cell biological and biochemical methods, we demonstrate that intracellular inhibition of S1P-lyase impairs the degradation of APP and CTFs in lysosomal compartments and also decreases the activity of γ-secretase. Interestingly, the strong accumulation of APP and CTFs in S1P-lyase-deficient cells was reversed by selective mobilization of Ca(2+) from the endoplasmic reticulum or lysosomes. Intracellular accumulation of S1P also impairs maturation of cathepsin D and degradation of Lamp-2, indicating a general impairment of lysosomal activity. Together, these data demonstrate that S1P-lyase plays a critical role in the regulation of lysosomal activity and the metabolism of APP.


Subject(s)
Aldehyde-Lyases/drug effects , Amyloid beta-Protein Precursor/metabolism , Lysosomes/metabolism , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Calcium/metabolism , Cathepsin D/metabolism , HEK293 Cells , Humans , Lysophospholipids/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Mice , Proteolysis , Sphingosine/analogs & derivatives , Sphingosine/metabolism
15.
FASEB J ; 28(2): 978-97, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24165480

ABSTRACT

In Alzheimer's disease (AD), disturbed homeostasis of the proteases competing for amyloid precursor protein processing has been reported: a disintegrin and metalloproteinase 10 (ADAM10), the physiological α-secretase, is decreased in favor of the amyloid-ß-generating enzyme BACE-1. To identify transcription factors that modulate the expression of either protease, we performed a screening approach: 48 transcription factors significantly interfered with ADAM10/BACE-1-promoter activity. One selective inducer of ADAM10 gene expression is the X-box binding protein-1 (XBP-1). This protein regulates the unfolded protein-response pathway. We demonstrate that particularly the spliced XBP-1 variant dose dependently regulates ADAM10 expression, which can be synergistically enhanced by 100 nM insulin. Analysis of 2 different transgenic mouse models (APP/PS1 and 5xFAD) revealed that at early time points in pathology XBP-1 metabolism is induced. This is accompanied by a 2-fold augmented ADAM10 amount as compared with nontransgenic littermates (P=0.011). Along with aging of the mice, the system is counterregulated, and XBP-1 together with ADAM10 expression level decreased to ∼50% as compared with control animals. Analyses of expression levels in human AD brains showed that ADAM10 mRNA correlated with active XBP-1 (r=0.3120), but expression did not reach levels of healthy age-matched controls, suggesting deregulation of XBP-1 signaling. Our results demonstrate that XBP-1 is a driver of ADAM10 gene expression and that disturbance of this pathway might contribute to development or progression of AD.


Subject(s)
ADAM Proteins/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Transcription Factors/metabolism , Unfolded Protein Response/physiology , ADAM Proteins/genetics , ADAM10 Protein , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Humans , Membrane Proteins/genetics , Mice , Promoter Regions, Genetic/genetics , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Unfolded Protein Response/genetics , X-Box Binding Protein 1
16.
J Neurosci ; 33(41): 16072-87, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24107941

ABSTRACT

Amyloid-ß (Aß), major constituent of senile plaques in Alzheimer's disease (AD), is generated by proteolytic processing of the amyloid precursor protein (APP) by ß- and γ-secretase. Several lipids, especially cholesterol, are associated with AD. Phytosterols are naturally occurring cholesterol plant equivalents, recently been shown to cross the blood-brain-barrier accumulating in brain. Here, we investigated the effect of the most nutritional prevalent phytosterols and cholesterol on APP processing. In general, phytosterols are less amyloidogenic than cholesterol. However, only one phytosterol, stigmasterol, reduced Aß generation by (1) directly decreasing ß-secretase activity, (2) reducing expression of all γ-secretase components, (3) reducing cholesterol and presenilin distribution in lipid rafts implicated in amyloidogenic APP cleavage, and by (4) decreasing BACE1 internalization to endosomal compartments, involved in APP ß-secretase cleavage. Mice fed with stigmasterol-enriched diets confirmed protective effects in vivo, suggesting that dietary intake of phytosterol blends mainly containing stigmasterol might be beneficial in preventing AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Cholesterol/metabolism , Membrane Microdomains/metabolism , Phytosterols/pharmacology , Animals , Blotting, Western , Brain Chemistry , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Flame Ionization , Gas Chromatography-Mass Spectrometry , Humans , Male , Membrane Microdomains/chemistry , Membrane Microdomains/drug effects , Mice , Mice, Inbred C57BL , Phytosterols/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stigmasterol/pharmacology
17.
Cell Physiol Biochem ; 34(1): 92-110, 2014.
Article in English | MEDLINE | ID: mdl-24977484

ABSTRACT

BACKGROUND: Gangliosides were found to be associated with Alzheimer's disease (AD). Here we addressed a potential function of γ-secretase (presenilin) dependent cleavage of the amyloid-precursor-protein (APP) in the regulation of ganglioside de novo synthesis. METHODS: To identify a potential role of γ-secretase and APP in ganglioside de novo synthesis we used presenilin (PS) deficient and APP deficient cells and mouse brains, mutated PS as well as transgenic mice and AD post mortem brains. Changes in glucosylceramide synthase (GCS) activity were identified by incorporation of radiolabeled UDP-glucose in glucosylceramide, changes in gene expression via real-time PCR and Western blot analysis. Alterations in ganglioside levels were determined by thin layer chromatography and mass spectrometry. RESULTS: We found that PS and APP deficiency, in vitro and in vivo, resulted in increased GCS gene expression, elevated enzyme activity and thus increased glucosylceramide and total ganglioside level. Using a specific γ-secretase inhibitor revealed that PS proteolytic activity alters ganglioside homeostasis. By the use of mutated PS causing early onset AD in cell culture and transgenic mice we found that GCS is increased in AD, further substantiated by the use of AD post mortem brains, suffering from sporadic AD. CONCLUSION: APP processing regulates ganglioside de novo synthesis and is affected in AD.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Protein Precursor/metabolism , Glucosyltransferases/metabolism , Presenilins/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Female , Gangliosides/metabolism , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Presenilins/deficiency , Presenilins/genetics , Transfection
18.
Neurodegener Dis ; 13(2-3): 75-81, 2014.
Article in English | MEDLINE | ID: mdl-24192346

ABSTRACT

Ninety percent of the elderly population has a vitamin D hypovitaminosis, and several lines of evidence suggest that there might be a potential causal link between Alzheimer's disease (AD) and a non-sufficient supply with vitamin D. However, the mechanisms linking AD to vitamin D have not been completely understood. The aim of our study is to elucidate the impact of 25(OH) vitamin D3 on amyloid precursor protein processing in mice and N2A cells utilizing very moderate and physiological vitamin D hypovitaminosis in the range of 20-30% compared to wild-type mice. We found that already under such mild conditions, amyloid-ß peptide (Aß) is significantly increased, which is caused by an increased ß-secretase activity and BACE1 protein level. Additionally, neprilysin (NEP) expression is downregulated resulting in a decreased NEP activity further enhancing the effect of decreased vitamin D on the Aß level. In line with the in vivo findings, corresponding effects were found with N2A cells supplemented with 25(OH) vitamin D3. Our results further strengthen the link between AD and vitamin D3 and suggest that supplementation of vitamin D3 might have a beneficial effect in AD prevention.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Cholecalciferol/metabolism , Vitamin D Deficiency/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Vitamin D Deficiency/complications
19.
Cell Chem Biol ; 31(2): 265-283.e7, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-37972592

ABSTRACT

Reduced sulfatide level is found in Alzheimer's disease (AD) patients. Here, we demonstrate that amyloid precursor protein (APP) processing regulates sulfatide synthesis and vice versa. Different cell culture models and transgenic mice models devoid of APP processing or in particular the APP intracellular domain (AICD) reveal that AICD decreases Gal3st1/CST expression and subsequently sulfatide synthesis. In return, sulfatide supplementation decreases Aß generation by reducing ß-secretase (BACE1) and γ-secretase processing of APP. Increased BACE1 lysosomal degradation leads to reduced BACE1 protein level in endosomes. Reduced γ-secretase activity is caused by a direct effect on γ-secretase activity and reduced amounts of γ-secretase components in lipid rafts. Similar changes were observed by analyzing cells and mice brain samples deficient of arylsulfatase A responsible for sulfatide degradation or knocked down in Gal3st1/CST. In line with these findings, addition of sulfatides to brain homogenates of AD patients resulted in reduced γ-secretase activity. Human brain APP level shows a significant negative correlation with GAL3ST1/CST expression underlining the in vivo relevance of sulfatide homeostasis in AD.


Subject(s)
Alzheimer Disease , Mice , Animals , Humans , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Sulfoglycosphingolipids , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic
20.
Int J Mol Sci ; 14(3): 5879-98, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23485990

ABSTRACT

Alzheimer's disease (AD) is characterized by extracellular accumulation of amyloid-ß peptide (Aß), generated by proteolytic processing of the amyloid precursor protein (APP) by ß- and γ-secretase. Aß generation is inhibited when the initial ectodomain shedding is caused by α-secretase, cleaving APP within the Aß domain. Therefore, an increase in α-secretase activity is an attractive therapeutic target for AD treatment. APP and the APP-cleaving secretases are all transmembrane proteins, thus local membrane lipid composition is proposed to influence APP processing. Although several studies have focused on γ-secretase, the effect of the membrane lipid microenvironment on α-secretase is poorly understood. In the present study, we systematically investigated the effect of fatty acid (FA) acyl chain length (10:0, 12:0, 14:0, 16:0, 18:0, 20:0, 22:0, 24:0), membrane polar lipid headgroup (phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine), saturation grade and the FA double-bond position on α-secretase activity. We found that α-secretase activity is significantly elevated in the presence of FAs with short chain length and in the presence of polyunsaturated FAs, whereas variations in the phospholipid headgroups, as well as the double-bond position, have little or no effect on α-secretase activity. Overall, our study shows that local lipid membrane composition can influence α-secretase activity and might have beneficial effects for AD.

SELECTION OF CITATIONS
SEARCH DETAIL