Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
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
2.
Front Cell Dev Biol ; 10: 859052, 2022.
Article in English | MEDLINE | ID: mdl-35557938

ABSTRACT

Cellular lipid metabolism is tightly regulated and requires a sophisticated interplay of multiple subcellular organelles to adapt to changing nutrient supply. PEX19 was originally described as an essential peroxisome biogenesis factor that selectively targets membrane proteins to peroxisomes. Metabolic aberrations that were associated with compromised PEX19 functions, were solely attributed to the absence of peroxisomes, which is also considered the underlying cause for Zellweger Spectrum Disorders. More recently, however, it was shown that PEX19 also mediates the targeting of the VCP/P97-recuitment factor UBXD8 to the ER from where it partitions to lipid droplets (LDs) but the physiological consequences remained elusive. Here, we addressed the intriguing possibility that PEX19 coordinates the functions of the major cellular sites of lipid metabolism. We exploited the farnesylation of PEX19 and deciphered the organelle-specific functions of PEX19 using systems level approaches. Non-farnesylated PEX19 is sufficient to fully restore the metabolic activity of peroxisomes, while farnesylated PEX19 controls lipid metabolism by a peroxisome-independent mechanism that can be attributed to sorting a specific protein subset to LDs. In the absence of this PEX19-dependent LD proteome, cells accumulate excess triacylglycerols and fail to fully deplete their neutral lipid stores under catabolic conditions, highlighting a hitherto unrecognized function of PEX19 in controlling neutral lipid storage and LD dynamics.

3.
Cells ; 10(11)2021 10 29.
Article in English | MEDLINE | ID: mdl-34831163

ABSTRACT

The accumulation of amyloid ß-protein (Aß) is one of the major pathological hallmarks of Alzheimer's disease. Insulin-degrading enzyme (IDE), a zinc-metalloprotease, is a key enzyme involved in Aß degradation, which, in addition to Aß production, is critical for Aß homeostasis. Here, we demonstrate that saturated medium-chain fatty acids (MCFAs) increase total Aß degradation whereas longer saturated fatty acids result in an inhibition of its degradation, an effect which could not be detected in IDE knock-down cells. Further analysis of the underlying molecular mechanism revealed that MCFAs result in an increased exosomal IDE secretion, leading to an elevated extracellular and a decreased intracellular IDE level whereas gene expression of IDE was unaffected in dependence of the chain length. Additionally, MCFAs directly elevated the enzyme activity of recombinant IDE, while longer-chain length fatty acids resulted in an inhibited IDE activity. The effect of MCFAs on IDE activity could be confirmed in mice fed with a MCFA-enriched diet, revealing an increased IDE activity in serum. Our data underline that not only polyunsaturated fatty acids such as docosahexaenoic acid (DHA), but also short-chain fatty acids, highly enriched, for example in coconut oil, might be beneficial in preventing or treating Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Fatty Acids/metabolism , Insulysin/metabolism , Proteolysis , Animals , Biocatalysis , Cell Line , Mice, Inbred C57BL , Models, Biological
4.
Sci Rep ; 11(1): 15301, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34315969

ABSTRACT

Alzheimer's disease (AD) is a very frequent neurodegenerative disorder characterized by an accumulation of amyloid-ß (Aß). Acitretin, a retinoid-derivative and approved treatment for Psoriasis vulgaris, increases non-amyloidogenic Amyloid-Precursor-Protein-(APP)-processing, prevents Aß-production and elicits cognitive improvement in AD mouse models. As an unintended side effect, acitretin could result in hyperlipidemia. Here, we analyzed the impact of acitretin on the lipidome in brain and liver tissue in the 5xFAD mouse-model. In line with literature, triglycerides were increased in liver accompanied by increased PCaa, plasmalogens and acyl-carnitines, whereas SM-species were decreased. In brain, these effects were partially enhanced or similar but also inverted. While for SM and plasmalogens similar effects were found, PCaa, TAG and acyl-carnitines showed an inverse effect in both tissues. Our findings emphasize, that potential pharmaceuticals to treat AD should be carefully monitored with respect to lipid-homeostasis because APP-processing itself modulates lipid-metabolism and medication might result in further and unexpected changes. Moreover, deducing effects of brain lipid-homeostasis from results obtained for other tissues should be considered cautiously. With respect to acitretin, the increase in brain plasmalogens might display a further positive probability in AD-treatment, while other results, such as decreased SM, indicate the need of medical surveillance for treated patients.


Subject(s)
Acitretin/pharmacology , Alzheimer Disease/drug therapy , Brain/metabolism , Disease Models, Animal , Lipidomics , Liver/metabolism , Models, Biological , Alzheimer Disease/metabolism , Animals , Mice
5.
Nutrients ; 13(3)2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33671099

ABSTRACT

Methylxanthines (MTX) are purine derived xanthine derivatives. Whereas naturally occurring methylxanthines like caffeine, theophylline or theobromine are widely consumed in food, several synthetic but also non-synthetic methylxanthines are used as pharmaceuticals, in particular in treating airway constrictions. Besides the well-established bronchoprotective effects, methylxanthines are also known to have anti-inflammatory and anti-oxidative properties, mediate changes in lipid homeostasis and have neuroprotective effects. Known molecular mechanisms include adenosine receptor antagonism, phosphodiesterase inhibition, effects on the cholinergic system, wnt signaling, histone deacetylase activation and gene regulation. By affecting several pathways associated with neurodegenerative diseases via different pleiotropic mechanisms and due to its moderate side effects, intake of methylxanthines have been suggested to be an interesting approach in dealing with neurodegeneration. Especially in the past years, the impact of methylxanthines in neurodegenerative diseases has been extensively studied and several new aspects have been elucidated. In this review we summarize the findings of methylxanthines linked to Alzheimer´s disease, Parkinson's disease and Multiple Sclerosis since 2017, focusing on epidemiological and clinical studies and addressing the underlying molecular mechanisms in cell culture experiments and animal studies in order to assess the neuroprotective potential of methylxanthines in these diseases.


Subject(s)
Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/administration & dosage , Xanthines/administration & dosage , Alzheimer Disease/drug therapy , Alzheimer Disease/epidemiology , Animals , Caffeine/administration & dosage , Coffee/chemistry , Humans , Multiple Sclerosis/drug therapy , Multiple Sclerosis/epidemiology , Parkinson Disease/drug therapy , Parkinson Disease/epidemiology , Theobromine/administration & dosage , Theophylline/administration & dosage
6.
Cell Rep ; 34(11): 108844, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33730587

ABSTRACT

Store-operated Ca2+-entry (SOCE) regulates basal and receptor-triggered Ca2+ signaling with STIM proteins sensing the endoplasmic reticulum (ER) Ca2+ content and triggering Ca2+ entry by gating Orai channels. Although crucial for immune cells, STIM1's role in neuronal Ca2+ homeostasis is controversial. Here, we characterize a splice variant, STIM1B, which shows exclusive neuronal expression and protein content surpassing conventional STIM1 in cerebellum and of significant abundance in other brain regions. STIM1B expression results in a truncated protein with slower kinetics of ER-plasma membrane (PM) cluster formation and ICRAC, as well as reduced inactivation. In primary wild-type neurons, STIM1B is targeted by its spliced-in domain B to presynaptic sites where it converts classic synaptic depression into Ca2+- and Orai-dependent short-term synaptic enhancement (STE) at high-frequency stimulation (HFS). In conjunction with altered STIM1 splicing in human Alzheimer disease, our findings highlight STIM1 splicing as an important regulator of neuronal calcium homeostasis and of synaptic plasticity.


Subject(s)
Stromal Interaction Molecule 1/metabolism , Synapses/metabolism , Animals , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Exons/genetics , HEK293 Cells , Humans , Mice, Inbred C57BL , Neurons/metabolism , ORAI1 Protein/metabolism , Phenotype , Presynaptic Terminals/metabolism , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing/genetics , Signal Transduction , Stromal Interaction Molecule 1/chemistry , Stromal Interaction Molecule 1/genetics
7.
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
8.
Aging Cell ; 19(11): e13264, 2020 11.
Article in English | MEDLINE | ID: mdl-33128835

ABSTRACT

One of the major pathological hallmarks of Alzheimer´s disease (AD) is an accumulation of amyloid-ß (Aß) in brain tissue leading to formation of toxic oligomers and senile plaques. Under physiological conditions, a tightly balanced equilibrium between Aß-production and -degradation is necessary to prevent pathological Aß-accumulation. Here, we investigate the molecular mechanism how insulin-degrading enzyme (IDE), one of the major Aß-degrading enzymes, is regulated and how amyloid precursor protein (APP) processing and Aß-degradation is linked in a regulatory cycle to achieve this balance. In absence of Aß-production caused by APP or Presenilin deficiency, IDE-mediated Aß-degradation was decreased, accompanied by a decreased IDE activity, protein level, and expression. Similar results were obtained in cells only expressing a truncated APP, lacking the APP intracellular domain (AICD) suggesting that AICD promotes IDE expression. In return, APP overexpression mediated an increased IDE expression, comparable results were obtained with cells overexpressing C50, a truncated APP representing AICD. Beside these genetic approaches, also AICD peptide incubation and pharmacological inhibition of the γ-secretase preventing AICD production regulated IDE expression and promoter activity. By utilizing CRISPR/Cas9 APP and Presenilin knockout SH-SY5Y cells results were confirmed in a second cell line in addition to mouse embryonic fibroblasts. In vivo, IDE expression was decreased in mouse brains devoid of APP or AICD, which was in line with a significant correlation of APP expression level and IDE expression in human postmortem AD brains. Our results show a tight link between Aß-production and Aß-degradation forming a regulatory cycle in which AICD promotes Aß-degradation via IDE and IDE itself limits its own production by degrading AICD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Insulysin/metabolism , Alzheimer Disease/pathology , Humans , Signal Transduction
9.
Sci Rep ; 10(1): 9164, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32514053

ABSTRACT

The vanilloid capsaicin is a widely consumed spice, known for its burning and "hot" sensation through activation of TRPV1 ion-channels, but also known to decrease oxidative stress, inflammation and influence tau-pathology. Beside these positive effects, little is known about its effects on amyloid-precursor-protein (APP) processing leading to amyloid-ß (Aß), the major component of senile plaques. Treatment of neuroblastoma cells with capsaicinoids (24 hours, 10 µM) resulted in enhanced Aß-production and reduced Aß-degradation, leading to increased Aß-levels. In detailed analysis of the amyloidogenic-pathway, both BACE1 gene-expression as well as protein-levels were found to be elevated, leading to increased ß-secretase-activity. Additionally, γ-secretase gene-expression as well as activity was enhanced, accompanied by a shift of presenilin from non-raft to raft membrane-domains where amyloidogenic processing takes place. Furthermore, impaired Aß-degradation in presence of capsaicinoids is dependent on the insulin-degrading-enzyme, one of the major Aß-degrading-enzymes. Regarding Aß-homeostasis, no differences were found between the major capsaicinoids, capsaicin and dihydrocapsaicin, and a mixture of naturally derived capsaicinoids; effects on Ca2+-homeostasis were ruled out. Our results show that in respect to Alzheimer's disease, besides the known positive effects of capsaicinoids, pro-amyloidogenic properties also exist, enhancing Aß-levels, likely restricting the potential use of capsaicinoids as therapeutic substances in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Capsaicin/adverse effects , Alzheimer Disease/etiology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Cell Line, Tumor , Contraindications, Drug , Gene Expression , Humans , Neuroblastoma
10.
J Nutr Biochem ; 67: 123-137, 2019 05.
Article in English | MEDLINE | ID: mdl-30889441

ABSTRACT

A vast majority of the elderly population shows a mild to moderate vitamin D deficiency. Besides the well-known function of vitamin D, vitamin D receptor is also expressed in brain and is discussed to regulate several genes. However very little is known whether genes are regulated, associated with Alzheimer's disease (AD). Here we investigate 117 genes, known to be affected in AD, in mouse brain samples with a mild vitamin D hypovitaminosis comparable to the vitamin D status of the elderly population (20%-30% deficiency). The 117 genes include two positive controls, Nep and Park7, already known to be affected by both AD and vitamin D hypovitaminosis. The 25 most promising candidates were verified in a second independent mouse cohort, resulting in eleven genes further evaluated against three additional housekeeping genes. Three of the remaining eight significantly altered genes are involved in APP homeostasis (Snca, Nep, Psmb5), and each one gene in oxidative stress (Park7), inflammation (Casp4), lipid metabolism (Abca1), signal transduction (Gnb5) and neurogenesis (Plat). Our results tighten the link of vitamin D and AD and underline that vitamin D influences several genes also in brain, highlighting that a strong link not only to AD but also to other neurodegenerative diseases might exist.


Subject(s)
Alzheimer Disease/genetics , Brain/physiology , Vitamin D Deficiency/genetics , Animals , Female , Gene Expression Profiling , Inflammation/genetics , Lipid Metabolism/genetics , Mice, Inbred C57BL , Oxidative Stress/genetics , Vitamin D Deficiency/etiology
11.
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
12.
Neurobiol Aging ; 72: 62-71, 2018 12.
Article in English | MEDLINE | ID: mdl-30216939

ABSTRACT

Amyloid-ß (Aß) plaques are a prominent pathological hallmark of Alzheimer's disease (AD). They consist of aggregated Aß peptides, which are generated through sequential proteolytic processing of the transmembrane protein amyloid precursor protein (APP) and several Aß-associated factors. Efficient clearance of Aß from the brain is thought to be important to prevent the development and progression of AD. The ubiquitin-proteasome system (UPS) is one of the major pathways for protein breakdown in cells and it has been suggested that impaired UPS-mediated removal of protein aggregates could play an important role in the pathogenesis of AD. To study the effects of an impaired UPS on Aß pathology in vivo, transgenic APPSwe/PS1ΔE9 mice (APPPS1) were crossed with transgenic mice expressing mutant ubiquitin (UBB+1), a protein-based inhibitor of the UPS. Surprisingly, the APPPS1/UBB+1 crossbreed showed a remarkable decrease in Aß plaque load during aging. Further analysis showed that UBB+1 expression transiently restored PS1-NTF expression and γ-secretase activity in APPPS1 mice. Concurrently, UBB+1 decreased levels of ß-APP-CTF, which is a γ-secretase substrate. Although UBB+1 reduced Aß pathology in APPPS1 mice, it did not improve the behavioral deficits in these animals.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Behavior, Animal , Plaque, Amyloid/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Amyloid beta-Peptides/genetics , Animals , Disease Models, Animal , Mice , Mice, Transgenic
13.
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
14.
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
15.
Front Mol Neurosci ; 10: 63, 2017.
Article in English | MEDLINE | ID: mdl-28344547

ABSTRACT

Extracellular neuritic plaques, composed of aggregated amyloid-ß (Aß) peptides, are one of the major histopathological hallmarks of Alzheimer's disease (AD), a progressive, irreversible neurodegenerative disorder and the most common cause of dementia in the elderly. One of the most prominent risk factor for sporadic AD, carrying one or two aberrant copies of the apolipoprotein E (ApoE) ε4 alleles, closely links AD to lipids. Further, several lipid classes and fatty acids have been reported to be changed in the brain of AD-affected individuals. Interestingly, the observed lipid changes in the brain seem not only to be a consequence of the disease but also modulate Aß generation. In line with these observations, protective lipids being able to decrease Aß generation and also potential negative lipids in respect to AD were identified. Mechanistically, Aß peptides are generated by sequential proteolytic processing of the amyloid precursor protein (APP) by ß- and γ-secretase. The α-secretase appears to compete with ß-secretase for the initial cleavage of APP, preventing Aß production. All APP-cleaving secretases as well as APP are transmembrane proteins, further illustrating the impact of lipids on Aß generation. Beside the pathological impact of Aß, accumulating evidence suggests that Aß and the APP intracellular domain (AICD) play an important role in regulating lipid homeostasis, either by direct effects or by affecting gene expression or protein stability of enzymes involved in the de novo synthesis of different lipid classes. This review summarizes the current literature addressing the complex bidirectional link between lipids and AD and APP processing including lipid alterations found in AD post mortem brains, lipids that alter APP processing and the physiological functions of Aß and AICD in the regulation of several lipid metabolism pathways.

16.
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
17.
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
18.
PLoS One ; 11(10): e0164298, 2016.
Article in English | MEDLINE | ID: mdl-27788151

ABSTRACT

Sphingolipids and the derived gangliosides have critical functions in spermatogenesis, thus mutations in genes involved in sphingolipid biogenesis are often associated with male infertility. We have generated a transgenic mouse line carrying an insertion in the sphingomyelin synthase gene Sms1, the enzyme which generates sphingomyelin species in the Golgi apparatus. We describe the spermatogenesis defect of Sms1-/- mice, which is characterized by sloughing of spermatocytes and spermatids, causing progressive infertility of male homozygotes. Lipid profiling revealed a reduction in several long chain unsaturated phosphatidylcholins, lysophosphatidylcholins and sphingolipids in the testes of mutants. Multi-Spectral Optoacoustic Tomography indicated blood-testis barrier dysfunction. A supplementary diet of the essential omega-3 docosahexaenoic acid and eicosapentaenoic acid diminished germ cell sloughing from the seminiferous epithelium and restored spermatogenesis and fertility in 50% of previously infertile mutants. Our findings indicate that SMS1 has a wider than anticipated role in testis polyunsaturated fatty acid homeostasis and for male fertility.


Subject(s)
Fertility , Transferases (Other Substituted Phosphate Groups)/metabolism , Aging/physiology , Alternative Splicing , Animals , Epididymis/drug effects , Epididymis/metabolism , Fatty Acids, Omega-3/biosynthesis , Fatty Acids, Omega-3/pharmacology , Fertility/drug effects , Infertility, Male/enzymology , Lipid Metabolism/drug effects , Male , Mice , Mutagenesis, Insertional , Promoter Regions, Genetic/genetics , Spermatogenesis/drug effects , Testis/drug effects , Testis/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics
19.
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
20.
Curr Alzheimer Res ; 13(11): 1277-1289, 2016.
Article in English | MEDLINE | ID: mdl-27335034

ABSTRACT

Retinoic acid, the bioactive metabolite of beta-carotene or vitamin A, plays a pleiotropic, multifunctional role in vertebrate development. Studies in rodents revealed that a diet deficient in vitamin A results in a complex neonatal syndrome (the VAD syndrome), manifested in many organs. In humans, the function of retinoic acid (RA) extends into adulthood, where it has important roles in fertility, vision, and suppression of neoplastic growth. In recent years, it has also been suggested that retinoic acid might potentially act as a therapeutically relevant drug in attenuating or even preventing neurodegenerative diseases such as Alzheimer's disease (AD). Here, we report that VAD leads to an increase in A-beta peptide levels while only minor effects were observed on expression levels of the amyloid precursor protein (APP) processing proteinases in wild type mice. In line with these findings, rescue of hypovitaminosis reduced A-beta amount to baseline and induced sApp-alpha secretion in combination with an increase of alpha-secretase Adam10. By comparing retinoic acid treatment starting from a full nutrition status and a "VAD" situation in human neuroblastoma cells, we show that while intensities of differential gene expression were higher in replenished cells, a large overlap in AD-related, regulated genes was observed. Our data suggest that hypovitaminosis A can contribute to onset or progression of AD by increasing synthesis of A-beta peptides and that several AD-related genes such as ADAM10 or BDNF are regulated by retinoic acid. We suggest that dietary supplementation with retinoic acid derivatives is likely to have a beneficial effect on AD-pathology in individuals with hypovitaminosis and patients with normal vitamin A status.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Vitamin A Deficiency/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Acitretin/chemistry , Acitretin/pharmacology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Female , Gene Regulatory Networks/drug effects , Humans , Keratolytic Agents/pharmacology , Mice , Neuroblastoma/metabolism , Neurons/drug effects , Peptide Fragments/metabolism , Presenilin-2/metabolism , Rats, Wistar , Tretinoin/chemistry , Tretinoin/metabolism , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...