Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Epigenetics ; 15(8): 781-799, 2020 08.
Article in English | MEDLINE | ID: mdl-32019393

ABSTRACT

The Presenilin1 (PSEN1) gene encodes the catalytic peptide of the γ-secretase complex, a key enzyme that cleaves the amyloid-ß protein precursor (AßPP), to generate the amyloid-ß (Aß) peptides, involved in Alzheimer's Disease (AD). Other substrates of the γ-secretase, such as E-cadherin and Notch1, are involved in neurodevelopment and haematopoiesis. Gene-specific DNA methylation influences PSEN1 expression in AD animal models. Here we evaluated canonical and non-canonical cytosine methylation patterns of the PSEN1 5'-flanking during brain development and AD progression, in DNA extracted from the frontal cortex of AD transgenic mice (TgCRND8) and post-mortem human brain. Mapping CpG and non-CpG methylation revealed different methylation profiles in mice and humans. PSEN1 expression only correlated with DNA methylation in adult female mice. However, in post-mortem human brain, lower methylation, both at CpG and non-CpG sites, correlated closely with higher PSEN1 expression during brain development and in disease progression. PSEN1 methylation in blood DNA was significantly lower in AD patients than in controls. The present study is the first to demonstrate a temporal correlation between dynamic changes in PSEN1 CpG and non-CpG methylation patterns and mRNA expression during neurodevelopment and AD neurodegeneration. These observations were made possible by the use of an improved bisulphite methylation assay employing primers that are not biased towards non-CpG methylation. Our findings deepen the understanding of γ-secretase regulation and support the hypothesis that epigenetic changes can promote the pathophysiology of AD. Moreover, they suggest that PSEN1 DNA methylation in peripheral blood may provide a biomarker for AD.


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , DNA Methylation , Presenilin-1/genetics , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Animals , Brain/growth & development , Brain/pathology , CpG Islands , Female , Humans , Male , Mice , Presenilin-1/metabolism
2.
Article in English | MEDLINE | ID: mdl-31202182

ABSTRACT

The multifactorial nature of Late Onset Alzheimer's Disease (LOAD), the AD form of major relevance on epidemiological and social aspects, has driven the original investigation by LC-MS and top-down proteomics approach of the protein repertoire of the brain tissue of TgCRND8 model mice fed with a diet deficient in B vitamins. The analysis of the acid-soluble fraction of brain tissue homogenates identified a list of proteins and peptides, proteoforms and PTMs. In order to disclose possible modulations, their relative quantification in wild type and AD model mice under both B vitamin deficient and control diets was performed. The levels of metallothionein III, guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2 and brain acid soluble protein 1 showed statistically significant alterations depending on genotype, diet or both effects, respectively. Particularly, metallothionein III exhibited increased levels in TgCRND8 mice under B vitamin deficient diet with respect to wild type mice under both diets. Brain acid soluble protein 1 showed the opposite, revealing decreased levels in all diet groups of AD model mice with respect to wild type mice in control diet. Lower levels of brain acid soluble protein 1 were also observed in wild type mice under deficiency of B vitamins. These results, besides contributing to increase the knowledge of AD at molecular level, give new suggestions for deeply investigating metallothionein III and brain acid soluble protein 1 in AD.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Hyperhomocysteinemia/metabolism , Proteome/metabolism , Vitamin B Complex/analysis , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain Chemistry , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Chromatography, Liquid , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Female , Humans , Hyperhomocysteinemia/etiology , Hyperhomocysteinemia/genetics , Male , Mass Spectrometry , Metallothionein 3 , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proteome/chemistry , Proteome/genetics , Vitamin B Complex/metabolism
3.
Antioxidants (Basel) ; 6(4)2017 Sep 30.
Article in English | MEDLINE | ID: mdl-28973985

ABSTRACT

Recent evidence emphasizes the role of dysregulated one-carbon metabolism in Alzheimer's Disease (AD). Exploiting a nutritional B-vitamin deficiency paradigm, we have previously shown that PSEN1 and BACE1 activity is modulated by one-carbon metabolism, leading to increased amyloid production. We have also demonstrated that S-adenosylmethionine (SAM) supplementation contrasted the AD-like features, induced by B-vitamin deficiency. In the present study, we expanded these observations by investigating the effects of SAM and SOD (Superoxide dismutase) association. TgCRND8 AD mice were fed either with a control or B-vitamin deficient diet, with or without oral supplementation of SAM + SOD. We measured oxidative stress by lipid peroxidation assay, PSEN1 and BACE1 expression by Real-Time Polymerase Chain Reaction (PCR), amyloid deposition by ELISA assays and immunohistochemistry. We found that SAM + SOD supplementation prevents the exacerbation of AD-like features induced by B vitamin deficiency, showing synergistic effects compared to either SAM or SOD alone. SAM + SOD supplementation also contrasts the amyloid deposition typically observed in TgCRND8 mice. Although the mechanisms underlying the beneficial effect of exogenous SOD remain to be elucidated, our findings identify that the combination of SAM + SOD could be carefully considered as co-adjuvant of current AD therapies.

4.
Curr Alzheimer Res ; 14(7): 753-759, 2017.
Article in English | MEDLINE | ID: mdl-28176663

ABSTRACT

BACKGROUND: The GSK3ß has been associated to pathological functions in neurodegenerative diseases. This kinase is involved in hyperphosphorylation of microtubule-associated tau protein, leading to aggregation andformation of NFTs. It has clearly been shown that GSK3ß is regulated at posttranslational level: phosphorylation at Tyr216 activates kinase, while phosphorylation at Ser9 is essential to inhibit its activity. OBJECTIVES: At present, there are contradictory findings about the possibility that GSK3ß may be regulated at gene level. Previous data showed overexpression of GSK3ß mRNA in hypomethylating conditions, pointing out to the existence of epigenetic mechanisms responsible for GSK3ß gene regulation. Analysis of human GSK3ß promoter through bisulphite modification, both in neuroblastoma cells and in postmortem frontal cortex from AD patients (AD patients both at Braak stages I-II and at stages V-VI) , allowed us to characterize the methylation pattern of a putative CpG islands in human GSK3ß 5'- flanking region. RESULTS: The analysis evidenced overall hypomethylation of CpG and non-CpG cytosine residues both in cells and in human brain (AD patients and control subjects). We found that GSK3ß mRNA was overexpressed only in patients with initial AD, with no effect on the levels of the protein. On the other hand, we unexpectedly observed the decrease of the inactive GSK3ß in cortex from AD patients at Braak stages I-II, whereas considerable increase was observed in AD patients at stages V-VI compared to the control subjects. CONCLUSIONS: These results point out that GSK3ß hyperactivity, and then NFTs formation, could come into function at an early stage of the disease and then turn off at the last stages.


Subject(s)
Alzheimer Disease/pathology , DNA Methylation/physiology , Frontal Lobe/enzymology , Glycogen Synthase Kinase 3 beta/genetics , 14-3-3 Proteins/metabolism , Aged , Aged, 80 and over , Analysis of Variance , Cell Line, Tumor , Female , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Middle Aged , Neuroblastoma/pathology , Neurofibrillary Tangles/pathology , Phosphorylation , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Serine/metabolism
5.
J Neuropathol Exp Neurol ; 76(1): 27-31, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28053004

ABSTRACT

By means of functional genomics analysis, we recently described the mRNA expression profiles of various genes involved in the neuroinflammatory response in the brains of subjects with late-onset Alzheimer Disease (LOAD). Some of these genes, namely interleukin (IL)-1ß and IL-6, showed distinct expression profiles with peak expression during the first stages of the disease and control-like levels at later stages. IL-1ß and IL-6 genes are modulated by DNA methylation in different chronic and degenerative diseases; it is also well known that LOAD may have an epigenetic basis. Indeed, we and others have previously reported gene-specific DNA methylation alterations in LOAD and in related animal models. Based on these data, we studied the DNA methylation profiles, at single cytosine resolution, of IL-1ß and IL-6 5'-flanking region by bisulphite modification in the cortex of healthy controls and LOAD patients at 2 different disease stages: Braak I-II/A and Braak V-VI/C. Our analysis provides evidence that neuroinflammation in LOAD is associated with (and possibly mediated by) epigenetic modifications.


Subject(s)
Alzheimer Disease/metabolism , Cytokines/metabolism , DNA Methylation/physiology , Gene Expression Profiling/methods , Inflammation Mediators/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cytokines/genetics , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Humans , Male , Middle Aged , Promoter Regions, Genetic/physiology
6.
J Mol Neurosci ; 61(3): 359-367, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27866325

ABSTRACT

Recent evidence highlights the protective role of reelin against amyloid ß (Aß)-induced synaptic dysfunction and cognitive impairment in Alzheimer disease (AD). In this study, exploiting TgCRND8 mice that overexpress a mutant form of amyloid ß precursor protein (AßPP) and display an early onset of AD neuropathological signs, we addressed the question whether changes of reelin expression eventually precede the appearance of Aß-plaques in a sex-dependent manner. We show that sex-associated and brain region-specific differences in reelin expression appear long before Aß-plaque formation. However, in spite of a downregulation of reelin expression compared to males, TgCRND8 females display fewer Aß-plaques, suggesting that additional factors, other than sex and reelin level, influence amyloidosis in this mouse model.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Cell Adhesion Molecules, Neuronal/genetics , Down-Regulation , Extracellular Matrix Proteins/genetics , Female , Male , Mice , Nerve Tissue Proteins/genetics , Organ Specificity , Reelin Protein , Serine Endopeptidases/genetics , Sex Factors
7.
J Alzheimers Dis ; 54(1): 307-24, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27567805

ABSTRACT

Amyloid-beta peptide accumulation in the brain is one of the main hallmarks of Alzheimer's disease. The amyloid aggregation process is associated with the generation of free radical species responsible for mitochondrial impairment and DNA damage that in turn activates poly(ADP-ribose)polymerase 1 (PARP-1). PARP-1 catalyzes the poly(ADP-ribosylation), a post-translational modification of proteins, cleaving the substrate NAD+ and transferring the ADP-ribose moieties to the enzyme itself or to an acceptor protein to form branched polymers of ADP-ribose. In this paper, we demonstrate that a mitochondrial dysfunction occurs in Alzheimer's transgenic mice TgCRND8, in SH-SY5Y treated with amyloid-beta and in 7PA2 cells. Moreover, PARP-1 activation contributes to the functional energetic decline affecting cytochrome oxidase IV protein levels, oxygen consumption rates, and membrane potential, resulting in cellular bioenergetic deficit. We also observed, for the first time, an increase of pyruvate kinase 2 expression, suggesting a modulation of the glycolytic pathway by PARP-1. PARP-1 inhibitors are able to restore both mitochondrial impairment and pyruvate kinase 2 expression. The overall data here presented indicate a pivotal role for this enzyme in the bioenergetic network of neuronal cells and open new perspectives for investigating molecular mechanisms underlying energy charge decline in Alzheimer's disease. In this scenario, PARP-1 inhibitors might represent a novel therapeutic intervention to rescue cellular energetic metabolism.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Neuroprotective Agents/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , CHO Cells , Cell Line, Tumor , Citrate (si)-Synthase/metabolism , Cricetulus , Disease Models, Animal , Electron Transport Complex IV/metabolism , Entorhinal Cortex/drug effects , Entorhinal Cortex/metabolism , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Lactic Acid/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , NAD/metabolism , Peptide Fragments/toxicity , Poly (ADP-Ribose) Polymerase-1/metabolism
8.
PLoS One ; 10(2): e0118318, 2015.
Article in English | MEDLINE | ID: mdl-25692551

ABSTRACT

Discordant results obtained in bisulfite assays using MethPrimers (PCR primers designed using MethPrimer software or assuming that non-CpGs cytosines are non methylated) versus primers insensitive to cytosine methylation lead us to hypothesize a technical bias. We therefore used the two kinds of primers to study different experimental models and methylation statuses. We demonstrated that MethPrimers negatively select hypermethylated DNA sequences in the PCR step of the bisulfite assay, resulting in CpG methylation underestimation and non-CpG methylation masking, failing to evidence differential methylation statuses. We also describe the characteristics of "Methylation-Insensitive Primers" (MIPs), having degenerated bases (G/A) to cope with the uncertain C/U conversion. As CpG and non-CpG DNA methylation patterns are largely variable depending on the species, developmental stage, tissue and cell type, a variable extent of the bias is expected. The more the methylome is methylated, the greater is the extent of the bias, with a prevalent effect of non-CpG methylation. These findings suggest a revision of several DNA methylation patterns so far documented and also point out the necessity of applying unbiased analyses to the increasing number of epigenomic studies.


Subject(s)
Alzheimer Disease/pathology , DNA Methylation , DNA Primers/analysis , Myogenin/genetics , Presenilin-1/genetics , Sequence Analysis, DNA/methods , Alzheimer Disease/genetics , Animals , Cell Line , CpG Islands , Humans , Mice , Software , Sulfites
9.
J Alzheimers Dis ; 44(4): 1323-31, 2015.
Article in English | MEDLINE | ID: mdl-25672765

ABSTRACT

Widely confirmed reports were published on association between hyperhomocysteinemia, B vitamin deficiency, oxidative stress, and amyloid-ß in Alzheimer's disease (AD). Homocysteine, cysteine, cysteinylglycine and glutathione are metabolically interrelated thiols that may be potential indicators of health status and disease risk; they all participate in the metabolic pathway of homocysteine. Previous data obtained in one of our laboratories showed that B vitamin deficiency induced exacerbation of AD-like features in TgCRND8 AD mice; these effects were counteracted by S-adenosylmethionine (SAM) supplementation, through the modulation of DNA methylation and antioxidant pathways. Since the cellular response to oxidative stress typically involves alteration in thiols content, a rapid and sensitive HPLC method with fluorescence detection was here used to evaluate the effect of SAM and superoxide-dismutase (SOD) supplementation on thiols level in plasma, in TgCRND8 mice. The quantitative data obtained from HPLC analysis of mice plasma samples showed significant decrease of thiols level when the B vitamin deficient diet was supplemented with SAM + SOD and SOD alone, the latter showing the greatest effect. All these considerations point out the measurement of plasma thiols concentration as a powerful tool of relevance for all clinical purposes involving the evaluation of oxidative stress. The coupling of HPLC with fluorimetric detection, here used, provided a strong method sensitivity allowing thiols determination at very low levels.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/diet therapy , Hyperhomocysteinemia/chemically induced , S-Adenosylmethionine/therapeutic use , Sulfhydryl Compounds/blood , Superoxide Dismutase/therapeutic use , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Chromatography , Chromatography, High Pressure Liquid , Disease Models, Animal , Glutathione/blood , Homocysteine/blood , Humans , Hyperhomocysteinemia/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics
10.
PLoS One ; 8(9): e72169, 2013.
Article in English | MEDLINE | ID: mdl-24086258

ABSTRACT

Amyloid beta peptide (Aß) causes neurodegeneration by several mechanisms including oxidative stress, which is known to induce DNA damage with the consequent activation of poly (ADP-ribose) polymerase (PARP-1). To elucidate the role of PARP-1 in the neurodegenerative process, SH-SY5Y neuroblastoma cells were treated with Aß25-35 fragment in the presence or absence of MC2050, a new PARP-1 inhibitor. Aß25-35 induces an enhancement of PARP activity which is prevented by cell pre-treatment with MC2050. These data were confirmed by measuring PARP-1 activity in CHO cells transfected with amylod precursor protein and in vivo in brains specimens of TgCRND8 transgenic mice overproducing the amyloid peptide. Following Aß25-35 exposure a significant increase in intracellular ROS was observed. These data were supported by the finding that Aß25-35 induces DNA damage which in turn activates PARP-1. Challenge with Aß25-35 is also able to activate NF-kB via PARP-1, as demonstrated by NF-kB impairment upon MC2050 treatment. Moreover, Aß25-35 via PARP-1 induces a significant increase in the p53 protein level and a parallel decrease in the anti-apoptotic Bcl-2 protein. These overall data support the hypothesis of PARP-1 involvment in cellular responses induced by Aß and hence a possible rationale for the implication of PARP-1 in neurodegeneration is discussed.


Subject(s)
Amyloid beta-Peptides/toxicity , Neurons/drug effects , Peptide Fragments/toxicity , Poly(ADP-ribose) Polymerases/physiology , Animals , Base Sequence , CHO Cells , Cell Line , Comet Assay , Cricetinae , Cricetulus , DNA Damage , DNA Primers , Electrophoretic Mobility Shift Assay , Mice , Mice, Transgenic , Poly (ADP-Ribose) Polymerase-1 , Reactive Oxygen Species/metabolism
11.
Behav Brain Res ; 237: 223-9, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23026376

ABSTRACT

Small GTPases of the Rho family, including Rho, Rac and CDC42 subfamilies, play key role in neural connectivity and cognition. The pharmacological modulation of these regulatory proteins is associated with enhancement of learning and memory. We sought to determine whether the modulation of cerebral Rho GTPases may correct behavioral disturbances in a mouse model of Alzheimer's disease (AD). TgCRND8 mice show early-onset Abeta amyloid deposits associated with deficits in several cognitive tasks. We report that four-month old TgCRND8 mice display (a) increased locomotor activity in an open field, (b) mild deficits in the learning of a fixed platform position in a water maze task. More markedly, after displacement of the escape platform, TgCRND8 mice exhibit impairment in the learning of the novel position (reversal learning), as they perseverate searching in the familiar position. The administration of the Rho GTPase activator Cytotoxic Necrotizing Factor 1 (CNF1, 1.0 fmol kg(-1) intracerebroventricularly) reduces locomotor hyperactivity and corrects the deficits in reversal learning, thus re-establishing normal behavioral plasticity. We conclude that the pharmacological modulation of Rho GTPase signaling might be beneficial for the treatment of AD. Reversal learning in TgCRND8 mice may represent a convenient pre-clinical assay for the efficacy of therapeutic interventions in AD.


Subject(s)
Alzheimer Disease/complications , Enzyme Activators/therapeutic use , Learning Disabilities/enzymology , Learning Disabilities/etiology , rho GTP-Binding Proteins/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Bacterial Toxins/therapeutic use , Disease Models, Animal , Escherichia coli Proteins/therapeutic use , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Humans , Learning Disabilities/drug therapy , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Mutation/genetics , Rotarod Performance Test
12.
Neurobiol Aging ; 33(7): 1482.e1-16, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22221883

ABSTRACT

Methylation reactions linked to homocysteine in the one-carbon metabolism are increasingly elicited in Alzheimer's disease, although the association of hyperhomocysteinemia and of low B vitamin levels with the disease is still debated. We previously demonstrated that hyperhomocysteinemia and DNA hypomethylation induced by B vitamin deficiency are associated with PSEN1 and BACE1 overexpression and amyloid production. The present study is aimed at assessing S-adenosylmethionine effects in mice kept under a condition of B vitamin deficiency. To this end, TgCRND8 mice and wild-type littermates were assigned to control or B vitamin deficient diet, with or without S-adenosylmethionine supplementation. We found that S-adenosylmethionine reduced amyloid production, increased spatial memory in TgCRND8 mice and inhibited the upregulation of B vitamin deficiency-induced PSEN1 and BACE1 expression and Tau phosphorylation in TgCRND8 and wild-type mice. Furthermore, S-adenosylmethionine treatment reduced plaque spreading independently on B vitamin deficiency. These results strengthen our previous observations on the possible role of one-carbon metabolism in Alzheimer's disease, highlighting hyperhomocysteinemia-related mechanisms in dementia onset/progression and encourage further studies aimed at evaluating the use of S-adenosylmethionine as a potential candidate drug for the treatment of the disease.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Disease Progression , S-Adenosylmethionine/therapeutic use , Vitamin B Deficiency/drug therapy , Vitamin B Deficiency/genetics , Action Potentials/physiology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/genetics , Vitamin B Deficiency/pathology
13.
Curr Alzheimer Res ; 9(5): 527-35, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22272624

ABSTRACT

In recent years, in parallel with the growing awareness of the multifactorial nature of Late Onset Alzheimer's Disease, the possibility that epigenetic mechanisms could be involved in the onset and/or progression of the pathology assumed an increasingly intriguing and leading role in Alzheimer's research. Today, many scientific reports indicate the existence of an epigenetic drift during ageing, in particular in Alzheimer's subjects. At the same time, experimental evidences are provided with the aim to demonstrate the causative or consequential role of epigenetic mechanisms. Our research group was involved in the last ten years in studying DNA methylation, the main epigenetic modification, in relationship to altered one-carbon metabolism (namely high homocysteine and low B vitamins levels), in Alzheimer's experimental models. Our previous findings about the demethylation of Presenilin1 gene promoter in nutritionally-induced hyperhomocysteinemia in a transgenic mouse model clearly demonstrated that Presenilin1 is regulated by DNA methylation. One of the open questions raised by our studies was if the observed demethylation was solely due to the induced imbalance of one-carbon metabolism or could be a response to the massive deposition of amyloid plaques in transgenic mice. Here we analyzed old (10 months) mice under standard diet in order to evidence possible changes in Presenilin1 promoter methylation in transgenic (TgCRND8 mice, carrying a double-mutated human APP transgene) vs. wt mice (129Sv) after prolonged exposure to amyloid. We found no differences in Presenilin1 methylation despite a slight increase in gene expression; these results suggest that amyloid production is not responsible for Presenilin1 demethylation in TgCRND8 mice brain.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , DNA Methylation , Presenilin-1/metabolism , Promoter Regions, Genetic , Aging/genetics , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Epigenesis, Genetic , Female , Gene Expression Regulation/physiology , Genetic Drift , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/metabolism , Male , Mice , Mice, 129 Strain , Mice, Transgenic , Presenilin-1/genetics
14.
Neurobiol Aging ; 32(7): 1192-5, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21524430

ABSTRACT

The sporadic form of Alzheimer disease, late onset Alzheimer's disease (LOAD), is a multifactorial disease; a strong link between nutritional and genetic factors with normal aging and dementia is supported by studies on nutrition, metabolism, and neurodegeneration. Specifically, the involvement of homocysteine (HCY) and its dietary determinants (vitamins B6, B12, and folate, besides methionine) in dementia has been a topic of intense investigation. In this Commentary we would like to highlight the role of 1-carbon metabolism in epigenetics and Alzheimer's disease and evidence the co-involvement of this metabolism in amyloid and tau pathways.


Subject(s)
Aging/genetics , Alzheimer Disease/genetics , Epigenesis, Genetic/physiology , Genetic Predisposition to Disease/genetics , Humans
15.
J Nutr Biochem ; 22(3): 242-51, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20573497

ABSTRACT

Late-onset Alzheimer's disease seems to be a multi-factorial disease with both genetic and non-genetic, environmental, possible causes. Recently, epigenomics is achieving a major role in Alzheimer's research due to its involvement in different molecular pathways leading to neurodegeneration. Among the different epigenetic modifications, DNA methylation is one of the most relevant to the disease. We previously demonstrated that presenilin1 (PSEN1), a gene involved in amyloidogenesis, is modulated by DNA methylation in neuroblastoma cells and Alzheimer's mice in an experimental model of nutritionally altered one-carbon metabolism. This alteration, obtained by nutritional deficiency of B vitamins (folate, B12 and B6) hampered S-adenosylmethionine (SAM)-dependent methylation reactions. The aim of the present paper was to investigate the regulation of DNA methylation machinery in response to hypomethylating (B vitamin deficiency) and hypermethylating (SAM supplementation) alterations of the one-carbon metabolism. We found that DNA methylases (DNMT1, 3a and 3b) and a putative demethylase (MBD2) were differently modulated, in line with the previously observed changes of PSEN1 methylation pattern in the same experimental conditions.


Subject(s)
Alzheimer Disease/metabolism , Carbon/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Disease Models, Animal , Analysis of Variance , Animals , Cell Line , Epigenomics , Female , Folic Acid/metabolism , Humans , Male , Methylation , Mice , Mice, 129 Strain , Mice, Transgenic , S-Adenosylmethionine/analysis , S-Adenosylmethionine/metabolism , Vitamin B 12/metabolism , Vitamin B 6/metabolism , Vitamin B Complex/metabolism , Vitamin B Complex/therapeutic use , Vitamin B Deficiency/metabolism
16.
Neurobiol Aging ; 32(2): 187-99, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19329227

ABSTRACT

We have previously shown that a nutritional model of B vitamin deficiency and homocysteine cycle alteration could lead to increased amyloid ß deposition, due to PSEN1 and BACE over-expression and consequent increase in secretase activity. We hypothesize that nutritional factors causing homocysteine cycle alterations (i.e. hyperhomocysteinemia) could induce sequence-specific DNA hypomethylation and "aberrant" gene activation. Aim of present study was to analyze the methylation pattern of PSEN1 promoter in SK-N-BE neuroblastoma cells and TgCRND8 mice, in a B vitamin (folate, B12 and B6) deficiency paradigm. PSEN1 methylation status has been evaluated through bisulphite modification and genomic sequencing. We demonstrate that B vitamin deficiency induces hypomethylation of specific CpG moieties in the 5'-flanking region; S-adenosylmethionine has been supplemented as methyl donor to reverse this effect. PSEN1 promoter methylation status is correlated with gene expression. These findings pinpoint a direct relationship between B vitamin-dependent alteration of homocysteine cycle and DNA methylation and also indicate that PSEN1 promoter is regulated by methylation of specific CpG moieties.


Subject(s)
DNA Methylation/physiology , Gene Expression Regulation/physiology , Presenilin-1/genetics , S-Adenosylmethionine/adverse effects , Vitamin B Deficiency/etiology , Vitamin B Deficiency/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Brain/metabolism , Cell Line, Tumor , DNA Methylation/drug effects , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Mice , Mice, Transgenic , Mutation/genetics , Presenilin-1/metabolism , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Sulfites/pharmacology , Transfection/methods , Vitamin B Deficiency/genetics , Vitamin B Deficiency/pathology
17.
Cell Cycle ; 9(19): 3965-76, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20935518

ABSTRACT

The dynamic changes and structural patterns of DNA methylation of genes without CpG islands are poorly characterized. The relevance of CpG to the non-CpG methylation equilibrium in transcriptional repression is unknown. In this work, we analyzed the DNA methylation pattern of the 5'-flanking of the myogenin gene, a positive regulator of muscle differentiation with no CpG island and low CpG density, in both C2C12 muscle satellite cells and embryonic muscle. Embryonic brain was studied as a non-expressing tissue. High levels of both CpG and non-CpG methylation were observed in non-expressing experimental conditions. Both CpG and non-CpG methylation rapidly dropped during muscle differentiation and myogenin transcriptional activation, with an active demethylation dynamics. Non-CpG demethylation occurred more rapidly than CpG demethylation. Demethylation spread from initially highly methylated short CpC-rich elements to a virtually unmethylated status. These short elements have a high CpC content and density, share some motifs and largely coincide with putative recognition sequences of some differentiation-related transcription factors. Our findings point to a dynamically controlled equilibrium between CpG and non-CpG active demethylation in the transcriptional control of tissue-specific genes. The short CpC-rich elements are new structural features of the methylation machinery, whose functions may include priming the complete demethylation of a transcriptionally crucial DNA region.


Subject(s)
5' Flanking Region/genetics , CpG Islands , DNA Methylation , Myogenin/genetics , Animals , Base Sequence , Cell Line , Cluster Analysis , Gene Expression Regulation , Humans , Mice , Molecular Sequence Data , Muscle Development/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Myogenin/metabolism , Promoter Regions, Genetic
18.
J Alzheimers Dis ; 22(4): 1209-21, 2010.
Article in English | MEDLINE | ID: mdl-20930312

ABSTRACT

The major mechanism of brain cholesterol elimination is the conversion of cholesterol into 24S-hydroxycholesterol by CYP46A1, a neuron-specific cytochrome P450. Since increasing evidence suggests that upregulation of CYP46A1 may be relevant for the treatment of Alzheimer's disease, we aim to identify the molecular mechanisms involved in CYP46A1 transcription. Our previous studies demonstrated the role of Sp transcription factors in basal expression and histone deacetylase (HDAC) inhibitor-dependent derepression of CYP46A1. Here, we show that the demethylating agent 5'-Aza-2'-deoxycytidine (DAC) is a CYP46A1 inducer and that pre-treatment with DAC causes a marked synergistic activation of CYP46A1 transcription by trichostatin A. Surprisingly, bisulfite sequencing analysis revealed that the CYP46A1 core promoter is completely unmethylated in both human brain and non-neuronal human tissues where CYP46A1 is not expressed. Therefore, we have investigated Sp expression levels by western blot and real-time PCR, and their binding patterns to the CYP46A1 promoter, by electrophoretic mobility shift assay and chromatin immunoprecipitation assays, after DAC treatment. Our results showed that DAC decreases not only Sp1 and Sp3 protein levels, but also the binding activity of Sp3 to the +1 region of the CYP46A1 locus. Concomitantly, HDAC1 and HDAC2 were also significantly dissociated from the promoter. In conclusion, DAC induces CYP46A1 gene expression, in a DNA methylation-independent mechanism, decreasing Sp3/HDAC binding to the proximal promoter. Furthermore, by affecting the expression of the Sp3 transcription factor in neuronal cells, DAC might affect not only brain cholesterol metabolism, but also the expression of many other neuronal genes.


Subject(s)
Azacitidine/analogs & derivatives , Cholesterol/metabolism , DNA Modification Methylases/antagonists & inhibitors , Neurons/drug effects , Steroid Hydroxylases/genetics , Analysis of Variance , Azacitidine/pharmacology , Blotting, Western , Cholesterol 24-Hydroxylase , Decitabine , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , HEK293 Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Neurons/metabolism , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Steroid Hydroxylases/metabolism
19.
J Alzheimers Dis ; 22(4): 1257-68, 2010.
Article in English | MEDLINE | ID: mdl-20930296

ABSTRACT

Late Onset Alzheimer's Disease (LOAD) can be associated to high homocysteine level and alteration of one-carbon metabolism. We previously demonstrated in the TgCRND8 mice strain, over-expressing human amyloid-ß protein precursor, that B vitamin deficiency causes alteration of one-carbon metabolism, together with unbalance of S-adenosylmethionine/S-adenosylhomocysteine levels, and is associated with AD like hallmarks as increased amyloid-ß plaque deposition, hyperhomocysteinemia, and oxidative stress. The same model of nutritional deficit was used here to study the variation of the brain protein expression profile associated to B vitamin deficiency. A group of proteins mainly involved in neuronal plasticity and mitochondrial functions was identified as modulated by one-carbon metabolism. These findings are consistent with increasing data about the pivotal role of mitochondrial abnormalities in AD patho-physiology. The identified proteins might represent new potential biomarkers of LOAD to be further investigated.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Carbon/metabolism , Proteome/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Cluster Analysis , Diet , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Female , Male , Mice , Mice, Transgenic , Neurons/metabolism , Oxidative Stress/physiology , Vitamin B Deficiency/genetics , Vitamin B Deficiency/metabolism
20.
J Alzheimers Dis ; 20(4): 997-1002, 2010.
Article in English | MEDLINE | ID: mdl-20413874

ABSTRACT

Oxidative stress, altered glutathione levels, and hyperhomocysteinemia play critical roles in Alzheimer's disease. We studied the relationships between hyperhomocysteinemia, glutathione, and oxidative stress in TgCRND8 mice maintained in conditions of folate, B12, and B6 deficiency and the effect of S-adenosylmethionine supplementation. We found that hyperhomocysteinemia was correlated with increased reduced/oxidized brain glutathione ratio, with decreased glutathione S-transferase activity and increased lipid peroxidation. S-adenosylmethionine potentiated superoxide dismutase and glutathione S-transferase activity and restored altered brain glutathione and erythrocytes lipid peroxidation. These results underline the importance of S-adenosylmethionine as neuroprotective compound, acting both on methylation and oxidation metabolism.


Subject(s)
Glutathione/metabolism , Oxidative Stress/drug effects , S-Adenosylmethionine/pharmacology , Vitamin B Deficiency/genetics , Vitamin B Deficiency/metabolism , Animals , Diet , Dietary Supplements , Erythrocytes/drug effects , Erythrocytes/metabolism , Glutathione Transferase/metabolism , Homocysteine/metabolism , Lipid Peroxidation/drug effects , Methylation , Mice , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...