Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(24)2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36555176

RESUMO

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.


Assuntos
Acitretina , Hiperlipidemias , Adulto , Humanos , Criança , Adolescente , Animais , Camundongos , Adulto Jovem , Acitretina/farmacologia , Acitretina/uso terapêutico , Lipidômica , Hiperlipidemias/induzido quimicamente , Colesterol , Encéfalo
2.
Cell Mol Life Sci ; 76(5): 1005-1025, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30599067

RESUMO

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.


Assuntos
Proteína ADAM10/genética , Doença de Alzheimer/etiologia , Regulação da Expressão Gênica , Proteínas com Domínio T/fisiologia , Secretases da Proteína Precursora do Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/genética , Sítios de Ligação , Encéfalo/metabolismo , Células Cultivadas , Desintegrinas/genética , Drosophila , Proteínas de Drosophila/genética , Histona Desacetilase 1/fisiologia , Humanos , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Regiões Promotoras Genéticas , Proteínas com Domínio T/química , Transcrição Gênica
3.
Int J Mol Sci ; 21(23)2020 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-33260941

RESUMO

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.


Assuntos
Doença de Alzheimer/genética , Cafeína/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Neuroblastoma/genética , Xantinas/farmacologia , Linhagem Celular Tumoral , Genes Essenciais , Humanos , Pentoxifilina/farmacologia , Análise de Componente Principal , Teobromina/farmacologia , Teofilina/farmacologia , Transcrição Gênica/efeitos dos fármacos , Xantinas/química
4.
J Lipid Res ; 58(11): 2083-2101, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28528321

RESUMO

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.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Apolipoproteínas E/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Alimentos , Doença de Alzheimer/dietoterapia , Animais , Humanos
5.
Int J Mol Sci ; 18(12)2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29257109

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Placa Amiloide/tratamento farmacológico , Proteólise , Vitamina D/uso terapêutico , Vitaminas/uso terapêutico , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Vitamina D/administração & dosagem , Vitamina D/farmacologia , Vitaminas/administração & dosagem , Vitaminas/farmacologia
6.
Biochem Cell Biol ; 94(6): 534-542, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27813426

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Insulisina/genética , Neuroblastoma/metabolismo , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Insulisina/metabolismo , Camundongos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , Neuroblastoma/patologia , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
7.
Ann Neurol ; 78(1): 88-103, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25893674

RESUMO

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.


Assuntos
ADP-Ribosil Ciclase 1/genética , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Comportamento Animal , Encéfalo/patologia , Glicoproteínas de Membrana/genética , Placa Amiloide/patologia , RNA Mensageiro/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microglia/metabolismo , Placa Amiloide/metabolismo , Aprendizagem Espacial , Transcriptoma
8.
Neurodegener Dis ; 16(1-2): 44-54, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26642316

RESUMO

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.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Ácidos Docosa-Hexaenoicos/análogos & derivados , Ácidos Docosa-Hexaenoicos/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/metabolismo , Animais , Membrana Celular/metabolismo , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Peroxidação de Lipídeos , Masculino , Espectrometria de Massas , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxirredução , Reação em Cadeia da Polimerase em Tempo Real , Bancos de Tecidos
9.
Int J Mol Sci ; 17(11)2016 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-27792188

RESUMO

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.


Assuntos
Doença de Alzheimer/metabolismo , Vitamina A/metabolismo , Vitamina D/metabolismo , Vitamina E/metabolismo , Vitamina K/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Metabolismo dos Lipídeos
10.
Int J Mol Sci ; 17(11)2016 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-27801864

RESUMO

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.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Neuroblastoma/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/biossíntese , Antioxidantes/administração & dosagem , Linhagem Celular , Colesterol/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Tocotrienóis/administração & dosagem , Vitamina E/administração & dosagem
11.
J Neurosci ; 33(41): 16072-87, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-24107941

RESUMO

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.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Colesterol/metabolismo , Microdomínios da Membrana/metabolismo , Fitosteróis/farmacologia , Animais , Western Blotting , Química Encefálica , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Ionização de Chama , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Masculino , Microdomínios da Membrana/química , Microdomínios da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fitosteróis/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estigmasterol/farmacologia
12.
Cell Physiol Biochem ; 34(1): 92-110, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24977484

RESUMO

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.


Assuntos
Doença de Alzheimer/enzimologia , Precursor de Proteína beta-Amiloide/metabolismo , Glucosiltransferases/metabolismo , Presenilinas/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Células COS , Linhagem Celular , Chlorocebus aethiops , Feminino , Gangliosídeos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Presenilinas/deficiência , Presenilinas/genética , Transfecção
13.
Neurodegener Dis ; 13(2-3): 75-81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24192346

RESUMO

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.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Colecalciferol/metabolismo , Deficiência de Vitamina D/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deficiência de Vitamina D/complicações
14.
Int J Mol Sci ; 14(3): 5879-98, 2013 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-23485990

RESUMO

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.

15.
J Biol Chem ; 286(16): 14028-39, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21324907

RESUMO

Alzheimer disease is characterized by accumulation of the ß-amyloid peptide (Aß) generated by ß- and γ-secretase processing of the amyloid precursor protein (APP). The intake of the polyunsaturated fatty acid docosahexaenoic acid (DHA) has been associated with decreased amyloid deposition and a reduced risk in Alzheimer disease in several epidemiological trials; however, the exact underlying molecular mechanism remains to be elucidated. Here, we systematically investigate the effect of DHA on amyloidogenic and nonamyloidogenic APP processing and the potential cross-links to cholesterol metabolism in vivo and in vitro. DHA reduces amyloidogenic processing by decreasing ß- and γ-secretase activity, whereas the expression and protein levels of BACE1 and presenilin1 remain unchanged. In addition, DHA increases protein stability of α-secretase resulting in increased nonamyloidogenic processing. Besides the known effect of DHA to decrease cholesterol de novo synthesis, we found cholesterol distribution in plasma membrane to be altered. In the presence of DHA, cholesterol shifts from raft to non-raft domains, and this is accompanied by a shift in γ-secretase activity and presenilin1 protein levels. Taken together, DHA directs amyloidogenic processing of APP toward nonamyloidogenic processing, effectively reducing Aß release. DHA has a typical pleiotropic effect; DHA-mediated Aß reduction is not the consequence of a single major mechanism but is the result of combined multiple effects.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Ácidos Docosa-Hexaenoicos/farmacologia , Proteínas ADAM/metabolismo , Proteína ADAM17 , Secretases da Proteína Precursora do Amiloide/metabolismo , Ração Animal , Animais , Ácido Aspártico Endopeptidases/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Colesterol/metabolismo , Hidroximetilglutaril-CoA Redutases/metabolismo , Lipídeos/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Presenilina-1/biossíntese , Risco
16.
Nat Cell Biol ; 7(11): 1118-23, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16227967

RESUMO

Amyloid beta peptide (Abeta) has a key role in the pathological process of Alzheimer's disease (AD), but the physiological function of Abeta and of the amyloid precursor protein (APP) is unknown. Recently, it was shown that APP processing is sensitive to cholesterol and other lipids. Hydroxymethylglutaryl-CoA reductase (HMGR) and sphingomyelinases (SMases) are the main enzymes that regulate cholesterol biosynthesis and sphingomyelin (SM) levels, respectively. We show that control of cholesterol and SM metabolism involves APP processing. Abeta42 directly activates neutral SMase and downregulates SM levels, whereas Abeta40 reduces cholesterol de novo synthesis by inhibition of HMGR activity. This process strictly depends on gamma-secretase activity. In line with altered Abeta40/42 generation, pathological presenilin mutations result in increased cholesterol and decreased SM levels. Our results demonstrate a biological function for APP processing and also a functional basis for the link that has been observed between lipids and Alzheimer's disease (AD).


Assuntos
Peptídeos beta-Amiloides/fisiologia , Precursor de Proteína beta-Amiloide/fisiologia , Colesterol/metabolismo , Metabolismo dos Lipídeos , Fragmentos de Peptídeos/análise , Esfingomielinas/metabolismo , Secretases da Proteína Precursora do Amiloide , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases , Células COS , Células Cultivadas , Chlorocebus aethiops , Endopeptidases/metabolismo , Regulação da Expressão Gênica , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Camundongos , Fragmentos de Peptídeos/metabolismo , Presenilina-1 , Presenilina-2 , Esfingomielina Fosfodiesterase/metabolismo
17.
Exp Brain Res ; 217(3-4): 365-75, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22179528

RESUMO

Amyloid plaques in brains are one of the major pathological hallmarks of Alzheimer's disease (AD). These plaques are mainly formed by aggregated Aß, generated by proteolytic cleavage of the amyloid precursor protein (APP). Therefore, APP processing and Aß production have been one of the central scopes in AD research in the past. Now, accumulating evidence suggests that besides its pathological impact, APP and its cleavage products also contribute to physiological functions. Proteolytic cleavage of APP is tightly regulated, and several lipids such as cholesterol and sphingolipids have been shown to influence APP processing and Aß generation. In turn, Aß as well as other APP cleavage products plays an essential role in regulating lipid homeostasis arguing for complex regulatory cycles in which lipids control APP processing and vice versa. This balanced regulation is disrupted under pathological conditions such as in AD. This article will review the physiological function of APP and its proteolytic products, especially Aß and AICD, in regulating lipid homeostasis and which lipid species modulate APP processing. Furthermore, we summarize the alterations in lipid metabolism observed in AD patients and AD mouse models.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Metabolismo dos Lipídeos/fisiologia , Proteólise , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Humanos , Processamento de Proteína Pós-Traducional/fisiologia
18.
ScientificWorldJournal ; 2012: 141240, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22547976

RESUMO

Lipids play an important role as risk or protective factors in Alzheimer's disease (AD). Previously it has been shown that plasmalogens, the major brain phospholipids, are altered in AD. However, it remained unclear whether plasmalogens themselves are able to modulate amyloid precursor protein (APP) processing or if the reduced plasmalogen level is a consequence of AD. Here we identify the plasmalogens which are altered in human AD postmortem brains and investigate their impact on APP processing resulting in Aß production. All tested plasmalogen species showed a reduction in γ-secretase activity whereas ß- and α-secretase activity mainly remained unchanged. Plasmalogens directly affected γ-secretase activity, protein and RNA level of the secretases were unaffected, pointing towards a direct influence of plasmalogens on γ-secretase activity. Plasmalogens were also able to decrease γ-secretase activity in human postmortem AD brains emphasizing the impact of plasmalogens in AD. In summary our findings show that decreased plasmalogen levels are not only a consequence of AD but that plasmalogens also decrease APP processing by directly affecting γ-secretase activity, resulting in a vicious cycle: Aß reduces plasmalogen levels and reduced plasmalogen levels directly increase γ-secretase activity leading to an even stronger production of Aß peptides.


Assuntos
Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Plasmalogênios/fisiologia , Processamento de Proteína Pós-Traducional , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Encéfalo/metabolismo , Linhagem Celular , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA/genética
19.
Front Cell Dev Biol ; 10: 859052, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35557938

RESUMO

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.

20.
Biochim Biophys Acta ; 1801(8): 966-74, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20452461

RESUMO

Alzheimer's disease (AD), the most common neurodegenerative disorder, which affects more than 35 million people worldwide, is characterized by a massive accumulation of tangles and amyloid plaques. Several risk factors linked to lipid homeostasis have been identified. Apolipoprotein E (ApoE), which also has a strong impact in coronary artery disease, is besides aging the most prominent risk factor in sporadic AD. High levels of lipoproteins and cholesterol increase the risk of AD and some cholesterol lowering drugs like statins seem to correlate with a reduced risk for dementia. Moreover, cholesterol increases amyloid beta (Abeta) production, which is derived from amyloid precursor protein (APP) by proteolytic processing. Beside cholesterol, other lipids that strongly modulate APP processing could be identified and interestingly the APP cleavage products itself regulate lipid homeostasis resulting in complex regulatory feedback cycles. Here, we review the mechanistic link of cholesterol and sphingolipid homeostasis and APP processing and the consequence of this bidirectional link for and in AD. Although cholesterol is the best studied brain lipid in AD, many other lipids are involved in the Abeta-lipid regulatory system and some of these lipids exceed the cholesterol effect on Abeta production [1-5]. This involvement is bidirectional. On the one hand, lipids control APP processing and, on the other hand, APP processing controls the levels of several key lipids [6, 7]. Beside the physiological function of APP processing in lipid homeostasis, under pathological conditions like AD, these regulating (feedback-) cycles are dysfunctional. Additionally, mutual influence of lipids and APP processing raises the question if altered lipid homeostasis is the cause or consequence of AD.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Metabolismo dos Lipídeos/fisiologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Secretases da Proteína Precursora do Amiloide/fisiologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Colesterol/metabolismo , Colesterol/fisiologia , Homeostase/fisiologia , Humanos , Lipídeos/fisiologia , Modelos Biológicos , Processamento de Proteína Pós-Traducional
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa