Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Alzheimers Dement ; 20(2): 1298-1308, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37985413

ABSTRACT

INTRODUCTION: Genome-wide association studies (GWAS) are fundamental for identifying loci associated with diseases. However, they require replication in other ethnicities. METHODS: We performed GWAS on sporadic Alzheimer's disease (AD) including 539 patients and 854 controls from Argentina and Chile. We combined our results with those from the European Alzheimer and Dementia Biobank (EADB) in a meta-analysis and tested their genetic risk score (GRS) performance in this admixed population. RESULTS: We detected apolipoprotein E ε4 as the single genome-wide significant signal (odds ratio  = 2.93 [2.37-3.63], P = 2.6 × 10-23 ). The meta-analysis with EADB summary statistics revealed four new loci reaching GWAS significance. Functional annotations of these loci implicated endosome/lysosomal function. Finally, the AD-GRS presented a similar performance in these populations, despite the score diminished when the Native American ancestry rose. DISCUSSION: We report the first GWAS on AD in a population from South America. It shows shared genetics modulating AD risk between the European and these admixed populations. HIGHLIGHTS: This is the first genome-wide association study on Alzheimer's disease (AD) in a population sample from Argentina and Chile. Trans-ethnic meta-analysis reveals four new loci involving lysosomal function in AD. This is the first independent replication for TREM2L, IGH-gene-cluster, and ADAM17 loci. A genetic risk score (GRS) developed in Europeans performed well in this population. The higher the Native American ancestry the lower the GRS values.


Subject(s)
Alzheimer Disease , Azides , Genome-Wide Association Study , Humans , Chile , Alzheimer Disease/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics
2.
Int J Mol Sci ; 24(23)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38069333

ABSTRACT

This work aims to clarify the effect of dietary polyunsaturated fatty acid (PUFA) intake on the adult brain affected by amyloid pathology. McGill-R-Thy1-APP transgenic (Tg) rat and 5xFAD Tg mouse models that represent earlier or later disease stages were employed. The animals were exposed to a control diet (CD) or an HFD based on corn oil, from young (rats) or adult (mice) ages for 24 or 10 weeks, respectively. In rats and mice, the HFD impaired reference memory in wild-type (WT) animals but did not worsen it in Tg, did not cause obesity, and did not increase triglycerides or glucose levels. Conversely, the HFD promoted stronger microglial activation in Tg vs. WT rats but had no effect on cerebral amyloid deposition. IFN-γ, IL-1ß, and IL-6 plasma levels were increased in Tg rats, regardless of diet, while CXCL1 chemokine levels were increased in HFD-fed mice, regardless of genotype. Hippocampal 3-nitrotyrosine levels tended to increase in HFD-fed Tg rats but not in mice. Overall, an HFD with an elevated omega-6-to-omega-3 ratio as compared to the CD (25:1 vs. 8.4:1) did not aggravate the outcome of AD regardless of the stage of amyloid pathology, suggesting that many neurobiological processes relevant to AD are not directly dependent on PUFA intake.


Subject(s)
Alzheimer Disease , Fatty Acids, Omega-3 , Mice , Rats , Animals , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Brain/pathology , Mice, Transgenic , Amyloid , Disease Models, Animal , Rats, Transgenic , Diet, High-Fat
3.
Article in English | MEDLINE | ID: mdl-36565982

ABSTRACT

Interest in the role of melanin-concentrating hormone (MCH) in memory processes has increased in recent years, with some studies reporting memory-enhancing effects, while others report deleterious effects. Due to these discrepancies, this study seeks to provide new evidence about the role of MCH in memory consolidation and its relation with BDNF/TrkB system. To this end, in the first experiment, increased doses of MCH were acutely administered in both hippocampi to groups of male rats (25, 50, 200, and 500 ng). Microinjections were carried out immediately after finishing the sample trial of two hippocampal-dependent behavioral tasks: the Novel Object Recognition Test (NORT) and the modified Elevated Plus Maze (mEPM) test. Results indicated that a dose of 200 ng of MCH or higher impaired memory consolidation in both tasks. A second experiment was performed in which a dose of 200 ng of MCH was administered alone or co-administered with the MCHR-1 antagonist ATC-0175 at the end of the sample trial in the NORT. Results showed that MCH impaired memory consolidation, while the co-administration with ATC-0175 reverted this detrimental effect. Moreover, MCH induced a significant decrease in hippocampal MCHR-1 and TrkB expression with no modification in the expression of BDNF and NMDA receptor subunits NR1, NR2A, and NR2B. These results suggest that MCH in vivo elicits pro-amnesic effects in the rat hippocampus by decreasing the availability of its receptor and TrkB receptors, thus linking both endogenous systems to memory processes.


Subject(s)
Brain-Derived Neurotrophic Factor , Memory Consolidation , Pituitary Hormones , Receptor, trkB , Receptors, Somatomedin , Animals , Male , Rats , Brain-Derived Neurotrophic Factor/metabolism , Melanins , Pituitary Hormones/metabolism , Receptor, trkB/metabolism , Receptors, Somatomedin/metabolism
4.
Front Mol Biosci ; 9: 1067296, 2022.
Article in English | MEDLINE | ID: mdl-36685284

ABSTRACT

Introduction: The metabolic routes altered in Alzheimer's disease (AD) brain are poorly understood. As the metabolic pathways are evolutionarily conserved, the metabolic profiles carried out in animal models of AD could be directly translated into human studies. Methods: We performed untargeted Nuclear Magnetic Resonance metabolomics in hippocampus of McGill-R-Thy1-APP transgenic (Tg) rats, a model of AD-like cerebral amyloidosis and the translational potential of these findings was assessed by targeted Gas Chromatography-Electron Impact-Mass Spectrometry in plasma of participants in the German longitudinal cohort AgeCoDe. Results: In rat hippocampus 26 metabolites were identified. Of these 26 metabolites, nine showed differences between rat genotypes that were nominally significant. Two of them presented partial least square-discriminant analysis (PLS-DA) loadings with the larger absolute weights and the highest Variable Importance in Projection (VIP) scores and were specifically assigned to nicotinamide adenine dinucleotide (NAD) and nicotinamide (Nam). NAD levels were significantly decreased in Tg rat brains as compared to controls. In agreement with these results, plasma of AD patients showed significantly reduced levels of Nam in respect to cognitively normal participants. In addition, high plasma levels of Nam showed a 27% risk reduction of progressing to AD dementia within the following 2.5 years, this hazard ratio is lost afterwards. Discussion: To our knowledge, this is the first report showing that a decrease of Nam plasma levels is observed couple of years before conversion to AD, thereby suggesting its potential use as biomarker for AD progression.

5.
Article in English | MEDLINE | ID: mdl-33042002

ABSTRACT

Several studies suggest that the assembly of mitochondrial respiratory complexes into structures known as supercomplexes (SCs) may increase the efficiency of the electron transport chain, reducing the rate of production of reactive oxygen species. Therefore, the study of the (dis)assembly of SCs may be relevant for the understanding of mitochondrial dysfunction reported in brain aging and major neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). Here we briefly reviewed the biogenesis and structural properties of SCs, the impact of mtDNA mutations and mitochondrial dynamics on SCs assembly, the role of lipids on stabilization of SCs and the methodological limitations for the study of SCs. More specifically, we summarized what is known about mitochondrial dysfunction and SCs organization and activity in aging, AD and PD. We focused on the critical variables to take into account when postmortem tissues are used to study the (dis)assembly of SCs. Since few works have been performed to study SCs in AD and PD, the impact of SCs dysfunction on the alteration of brain energetics in these diseases remains poorly understood. The convergence of future progress in the study of SCs structure at high resolution and the refinement of animal models of AD and PD, as well as the use of iPSC-based and somatic cell-derived neurons, will be critical in understanding the biological relevance of the structural remodeling of SCs.


Subject(s)
Brain/metabolism , Energy Metabolism/physiology , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Aging/metabolism , Aging/pathology , Animals , Brain/pathology , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Humans , Mitochondria/pathology , Neurodegenerative Diseases/pathology , Neurons/pathology , Reactive Oxygen Species/metabolism
6.
J Cell Sci ; 132(20)2019 10 22.
Article in English | MEDLINE | ID: mdl-31515277

ABSTRACT

The use of fixed fibroblasts from familial and sporadic Alzheimer's disease patients has previously indicated an upregulation of mitochondria-ER contacts (MERCs) as a hallmark of Alzheimer's disease. Despite its potential significance, the relevance of these results is limited because they were not extended to live neurons. Here we performed a dynamic in vivo analysis of MERCs in hippocampal neurons from McGill-R-Thy1-APP transgenic rats, a model of Alzheimer's disease-like amyloid pathology. Live FRET imaging of neurons from transgenic rats revealed perturbed 'lipid-MERCs' (gap width <10 nm), while 'Ca2+-MERCs' (10-20 nm gap width) were unchanged. In situ TEM showed no significant differences in the lipid-MERCs:total MERCs or lipid-MERCs:mitochondria ratios; however, the average length of lipid-MERCs was significantly decreased in neurons from transgenic rats as compared to controls. In accordance with FRET results, untargeted lipidomics showed significant decreases in levels of 12 lipids and bioenergetic analysis revealed respiratory dysfunction of mitochondria from transgenic rats. Thus, our results reveal changes in MERC structures coupled with impaired mitochondrial functions in Alzheimer's disease-related neurons.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Alzheimer Disease , Endoplasmic Reticulum , Mitochondria , Neurons , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Rats , Rats, Transgenic
7.
J Alzheimers Dis ; 68(2): 439-458, 2019.
Article in English | MEDLINE | ID: mdl-30775999

ABSTRACT

The accumulation and spreading of protein tau in the human brain are major features of neurodegenerative disorders known as tauopathies. In addition to several subcellular abnormalities, tau aggregation within neurons seems capable of triggering endoplasmic reticulum (ER) stress and the consequent unfolded protein response (UPR). In metazoans, full activation of a complex ER-UPR network may restore proteostasis and ER function or, if stress cannot be solved, commit cells to apoptosis. Due to these alternative outcomes (survival or death), the pharmacological manipulation of ER-UPR has become the focus of potential therapies in many human diseases, including tauopathies. Here we update and analyze the experimental data from human brain, cellular, and animal models linking tau accumulation and ER-UPR. We further discuss mechanistic aspects and put the ER-UPR into perspective as a possible therapeutic target in this group of diseases.


Subject(s)
Brain/metabolism , Brain/pathology , Endoplasmic Reticulum Stress , Tauopathies/metabolism , Tauopathies/pathology , Animals , Brain/drug effects , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Humans , Tauopathies/drug therapy
8.
Transl Psychiatry ; 9(1): 55, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30705288

ABSTRACT

Rare coding variants in TREM2, PLCG2, and ABI3 were recently associated with the susceptibility to Alzheimer's disease (AD) in Caucasians. Frequencies and AD-associated effects of variants differ across ethnicities. To start filling the gap on AD genetics in South America and assess the impact of these variants across ethnicity, we studied these variants in Argentinian population in association with ancestry. TREM2 (rs143332484 and rs75932628), PLCG2 (rs72824905), and ABI3 (rs616338) were genotyped in 419 AD cases and 486 controls. Meta-analysis with European population was performed. Ancestry was estimated from genome-wide genotyping results. All variants show similar frequencies and odds ratios to those previously reported. Their association with AD reach statistical significance by meta-analysis. Although the Argentinian population is an admixture, variant carriers presented mainly Caucasian ancestry. Rare coding variants in TREM2, PLCG2, and ABI3 also modulate susceptibility to AD in populations from Argentina, and they may have a European heritage.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/ethnology , Alzheimer Disease/genetics , Membrane Glycoproteins/genetics , Phospholipase C gamma/genetics , Receptors, Immunologic/genetics , Aged , Aged, 80 and over , Argentina/ethnology , Black People/genetics , Female , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Genotype , Humans , Indians, North American/genetics , Male , Middle Aged , White People/genetics
9.
Neurochem Res ; 44(6): 1375-1386, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30357651

ABSTRACT

Alzheimer's disease (AD) is associated to depressed brain energy supply and impaired cortical and hippocampal synaptic function. It was previously reported in McGill-R-Thy1-APP transgenic (Tg(+/+)) rats that Aß deposition per se is sufficient to cause abnormalities in glucose metabolism and neuronal connectivity. These data support the utility of this animal model as a platform for the search of novel AD biomarkers based on bioenergetic status. Recently, it has been proposed that energy dysfunction can be dynamically tested in platelets (PLTs) of nonhuman primates. PLTs are good candidates to find peripheral biomarkers for AD because they may reflect in periphery the bioenergetics deficits and the inflammatory and oxidative stress processes taking place in AD brain. In the present study, we carried out a PLTs bioenergetics screening in advanced-age (12-14 months old) control (WT) and Tg(+/+) rats. Results indicated that thrombin-activated PLTs of Tg(+/+) rats showed a significantly lower respiratory rate, as compared to that measured in WT animals, when challenged with the same dose of FCCP (an uncoupler of oxidative phosphorylation). In summary, our results provide original evidence that PLTs bioenergetic profiling may reflect brain bioenergetics dysfunction mediated by Aß plaque accumulation. Further studies on human PLTs from control and AD patients are required to validate the usefulness of PLTs bioenergetics as a novel blood-based biomarker for AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Blood Platelets/metabolism , Energy Metabolism/physiology , Hippocampus/metabolism , Plaque, Amyloid/metabolism , Animals , Mitochondria/metabolism , Rats , Rats, Transgenic
10.
Mol Neurobiol ; 55(11): 8637-8650, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29582397

ABSTRACT

The specific roles of Notch in progressive adulthood neurodegenerative disorders have begun to be unraveled in recent years. A number of independent studies have shown significant increases of Notch expression in brains from patients at later stages of sporadic Alzheimer's disease (AD). However, the impact of Notch canonical signaling activation in the pathophysiology of AD is still elusive. To further investigate this issue, 2-month-old wild-type (WT) and hemizygous McGill-R-Thy1-APP rats (Tg(+/-)) were injected in CA1 with lentiviral particles (LVP) expressing the transcriptionally active fragment of Notch, known as Notch Intracellular Domain (NICD), (LVP-NICD), or control lentivirus particles (LVP-C). The Tg(+/-) rat model captures presymptomatic aspects of the AD pathology, including intraneuronal amyloid beta (Aß) accumulation and early cognitive deficits. Seven months after LVP administration, Morris water maze test was performed, and brains isolated for biochemical and histological analysis. Our results showed a learning impairment and a worsening of spatial memory in LVP-NICD- as compared to LVP-C-injected Tg(+/-) rats. In addition, immuno histochemistry, ELISA multiplex, Western blot, RT-qPCR, and 1H-NMR spectrometry of cerebrospinal fluid (CSF) indicated that chronic expression of NICD promoted hippocampal vessel thickening with accumulation of Aß in brain microvasculature, alteration of blood-brain barrier (BBB) permeability, and a decrease of CSF glucose levels. These findings suggest that, in the presence of early Aß pathology, expression of NICD may contribute to the development of microvascular abnormalities, altering glucose transport at the BBB with impact on early decline of spatial learning and memory.


Subject(s)
Alzheimer Disease/pathology , Blood Vessels/pathology , Glucose/metabolism , Hippocampus/metabolism , Memory Disorders/pathology , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Spatial Memory , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/complications , Alzheimer Disease/physiopathology , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Disease Models, Animal , Genetic Vectors/metabolism , HEK293 Cells , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Inflammation/pathology , Lentivirus/genetics , Memory Disorders/complications , Memory Disorders/physiopathology , Microvessels/pathology , Protein Domains , Proton Magnetic Resonance Spectroscopy , Rats, Transgenic , Rats, Wistar
11.
Neurobiol Aging ; 60: 57-70, 2017 12.
Article in English | MEDLINE | ID: mdl-28917667

ABSTRACT

The unfolded protein response (UPR) may be pathogenically related to Alzheimer's disease. Yet, the effects of chronic amyloid-ß42 (Aß42) accumulation and UPR activation upon neurotoxicity remain unclear. Here, we show that neuronal Aß42 expression in Drosophila activated the inositol-requiring protein-1/X-box binding protein 1 (XBP1) UPR branch before the onset of behavioral impairment and persisted with aging. Early upregulation of hsc3/BiP, a target of XBP1 and activating transcription factor 6 pathways, was also sustained in old animals. Downregulation of XBP1 enhanced neurotoxicity and the accumulation of Aß42 and polyubiquitinated proteins. Consistently, overexpression of spliced XBP1 reduced Aß42 and improved geotaxis in old flies. The activation of protein kinase RNA-like endoplasmic reticulum (ER) kinase contributed to the age-dependent geotaxis deficit. Spliced XBP1 gene targets ER degradation-enhancing mannosidase-like protein 1, ER degradation-enhancing mannosidase-like protein 2, and HRD1 were elevated in 5-day-old Aß42-expressing flies as compared to controls but not in 18-day-old flies. Our results indicate that inositol-requiring protein-1/XBP1 activation is neuroprotective and enhances Aß42 clearance. They also suggest that such response becomes inefficient with aging.


Subject(s)
Aging , Amyloid beta-Peptides/metabolism , Brain/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Neuroprotection/genetics , Peptide Fragments/metabolism , Signal Transduction , X-Box Binding Protein 1/physiology , Animals
12.
Cell Rep ; 19(1): 72-85, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28380364

ABSTRACT

The small ventral lateral neurons (sLNvs) constitute a central circadian pacemaker in the Drosophila brain. They organize daily locomotor activity, partly through the release of the neuropeptide pigment-dispersing factor (PDF), coordinating the action of the remaining clusters required for network synchronization. Despite extensive efforts, the basic principles underlying communication among circadian clusters remain obscure. We identified classical neurotransmitters released by sLNvs through disruption of specific transporters. Adult-specific RNAi-mediated downregulation of the glycine transporter or impairment of glycine synthesis in LNv neurons increased period length by nearly an hour without affecting rhythmicity of locomotor activity. Electrophysiological recordings showed that glycine reduces spiking frequency in circadian neurons. Interestingly, downregulation of glycine receptor subunits in specific sLNv targets impaired rhythmicity, revealing involvement of glycine in information processing within the network. These data identify glycinergic inhibition of specific targets as a cue that contributes to the synchronization of the circadian network.


Subject(s)
Circadian Rhythm/physiology , Glycine Plasma Membrane Transport Proteins/metabolism , Glycine/metabolism , Receptors, Glycine/metabolism , Synaptic Transmission , Animals , Animals, Genetically Modified , Brain/metabolism , Down-Regulation , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Glycine Plasma Membrane Transport Proteins/genetics , Humans , Neurons/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , RNA Interference , Receptors, Glycine/genetics
13.
Front Aging Neurosci ; 9: 61, 2017.
Article in English | MEDLINE | ID: mdl-28352227

ABSTRACT

The accumulation of amyloid ß peptide (Aß) in the brain of Alzheimer's disease (AD) patients begins many years before clinical onset. Such process has been proposed to be pathogenic through the toxicity of Aß soluble oligomers leading to synaptic dysfunction, phospho-tau aggregation and neuronal loss. Yet, a massive accumulation of Aß can be found in approximately 30% of aged individuals with preserved cognitive function. Therefore, within the frame of the "amyloid hypothesis", compensatory mechanisms and/or additional neurotoxic or protective factors need to be considered and investigated. Here we describe a modifier genetic screen in Drosophila designed to identify genes that modulate toxicity of Aß42 in the CNS. The expression of Aß42 led to its accumulation in the brain and a moderate impairment of negative geotaxis at 18 days post-eclosion (d.p.e) as compared with genetic or parental controls. These flies were mated with a collection of lines carrying chromosomal deletions and negative geotaxis was assessed at 5 and 18 d.p.e. Our screen is the first to take into account all of the following features, relevant to sporadic AD: (1) pan-neuronal expression of wild-type Aß42; (2) a quantifiable complex behavior; (3) Aß neurotoxicity associated with progressive accumulation of the peptide; and (4) improvement or worsening of climbing ability only evident in aged animals. One hundred and ninety-nine deficiency (Df) lines accounting for ~6300 genes were analyzed. Six lines, including the deletion of 52 Drosophila genes with human orthologs, significantly modified Aß42 neurotoxicity in 18-day-old flies. So far, we have validated CG11796 and identified CG17249 as a strong candidate (whose human orthologs are HPD and PRCC, respectively) by using RNAi or mutant hemizygous lines. PRCC encodes proline-rich protein PRCC (ppPRCC) of unknown function associated with papillary renal cell carcinoma. HPD encodes 4-hydroxyphenylpyruvate dioxygenase (HPPD), a key enzyme in tyrosine degradation whose Df causes autosomal recessive Tyrosinemia type 3, characterized by mental retardation. Interestingly, lines with a partial Df of HPD ortholog showed increased intraneuronal accumulation of Aß42 that coincided with geotaxis impairment. These previously undetected modifiers of Aß42 neurotoxicity in Drosophila warrant further study to validate their possible role and significance in the pathogenesis of sporadic AD.

14.
Biochim Biophys Acta Mol Basis Dis ; 1863(3): 731-743, 2017 03.
Article in English | MEDLINE | ID: mdl-28039031

ABSTRACT

Diet is a modifiable risk factor for Alzheimer's disease (AD), but the mechanisms linking alterations in peripheral metabolism and cognition remain unclear. Since it is especially difficult to study long-term effects of high-energy diet in individuals at risk for AD, we addressed this question by using the McGill-R-Thy1-APP transgenic rat model (Tg(+/-)) that mimics presymptomatic AD. Wild-type and Tg(+/-) rats were exposed during 6months to a standard diet or a Western diet (WD), high in saturated fat and sugar. Results from peripheral and hippocampal biochemical analysis and in situ respirometry showed that WD induced a metabolic syndrome and decreased presynaptic bioenergetic parameters without alterations in hippocampal insulin signaling or lipid composition. Cognitive tests, ELISA multiplex, Western blot, immunohistochemistry and RT-qPCR indicated that WD worsened cognition in Tg(+/-) rats, increased hippocampal levels of monomeric Aß isoforms and oligomeric species, promoted deposits of N-Terminal pyroglutamate-Aß (AßN3(pE)) in CA1 pyramidal neurons and interneurons, decreased transcript levels of genes involved in neuroprotective pathways such as Sirtuin-1 and increased nitrated proteins. Our results support the concept that in the presence of early Aß pathology, diet-induced metabolic dysfunctions may contribute as a "second hit" to impair cognition. Noteworthy, such effect is not mediated by higher microglia activation or disruption of blood brain barrier. However, it may be attributed to increased amyloidogenic processing of amyloid precursor protein, generation of AßN3(pE) and dysregulation of pathways governed by Sirtuin-1. This evidence reinforces the implementation of prophylactic interventions in individuals at risk for AD.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Protein Precursor/metabolism , Diet, Western/adverse effects , Memory Disorders/etiology , Pyrrolidonecarboxylic Acid/metabolism , Adiposity , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cognition , Disease Models, Animal , Energy Metabolism , Gene Deletion , Hippocampus/metabolism , Humans , Inflammation/complications , Inflammation/genetics , Inflammation/metabolism , Male , Memory Disorders/metabolism , Rats , Rats, Transgenic
15.
J Cereb Blood Flow Metab ; 37(1): 69-84, 2017 01.
Article in English | MEDLINE | ID: mdl-26661224

ABSTRACT

Synaptic bioenergetic deficiencies may be associated with early Alzheimer's disease (AD). To explore this concept, we assessed pre-synaptic mitochondrial function in hemizygous (+/-)TgMcGill-R-Thy1-APP rats. The low burden of Aß and the wide array of behavioral and cognitive impairments described in 6-month-old hemizygous TgMcGill-R-Thy1-APP rats (Tg(+/-)) support their use to investigate synaptic bioenergetics deficiencies described in subjects with early Alzheimer's disease (AD). In this report, we show that pre-synaptic mitochondria from Tg(+/-) rats evidence a decreased respiratory control ratio and spare respiratory capacity associated with deficits in complex I enzymatic activity. Cognitive impairments were prevented and bioenergetic deficits partially reversed when Tg(+/-) rats were fed a nutritionally complete diet from weaning to 6-month-old supplemented with pyrroloquinoline quinone, a mitochondrial biogenesis stimulator with antioxidant and neuroprotective effects. These results provide evidence that, as described in AD brain and not proven in Tg mice models with AD-like phenotype, the mitochondrial bioenergetic capacity of synaptosomes is not conserved in the Tg(+/-) rats. This animal model may be suitable for understanding the basic biochemical mechanisms involved in early AD.


Subject(s)
Alzheimer Disease/etiology , Cognitive Dysfunction/etiology , Energy Metabolism , Synaptosomes/metabolism , Alzheimer Disease/diet therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cognitive Dysfunction/metabolism , Diet Therapy , Disease Models, Animal , Electron Transport Complex I/metabolism , Mitochondria/metabolism , PQQ Cofactor/therapeutic use , Rats , Rats, Transgenic
16.
PLoS Genet ; 10(10): e1004700, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25356918

ABSTRACT

In the Drosophila brain, the neuropeptide PIGMENT DISPERSING FACTOR (PDF) is expressed in the small and large Lateral ventral neurons (LNvs) and regulates circadian locomotor behavior. Interestingly, PDF immunoreactivity at the dorsal terminals changes across the day as synaptic contacts do as a result of a remarkable remodeling of sLNv projections. Despite the relevance of this phenomenon to circuit plasticity and behavior, the underlying mechanisms remain poorly understood. In this work we provide evidence that PDF along with matrix metalloproteinases (Mmp1 and 2) are key in the control of circadian structural remodeling. Adult-specific downregulation of PDF levels per se hampers circadian axonal remodeling, as it does altering Mmp1 or Mmp2 levels within PDF neurons post-developmentally. However, only Mmp1 affects PDF immunoreactivity at the dorsal terminals and exerts a clear effect on overt behavior. In vitro analysis demonstrated that PDF is hydrolyzed by Mmp1, thereby suggesting that Mmp1 could directly terminate its biological activity. These data demonstrate that Mmp1 modulates PDF processing, which leads to daily structural remodeling and circadian behavior.


Subject(s)
Circadian Rhythm/genetics , Drosophila Proteins/genetics , Matrix Metalloproteinase 1/genetics , Neuronal Plasticity/genetics , Neuropeptides/genetics , Animals , Animals, Genetically Modified , Behavior, Animal , Drosophila melanogaster , Motor Activity/genetics , Neurons/metabolism , Neurons/physiology
17.
Front Behav Neurosci ; 8: 321, 2014.
Article in English | MEDLINE | ID: mdl-25278855

ABSTRACT

Intraneuronal accumulation of amyloid ß (iAß) has been linked to mild cognitive impairment that may precede Alzheimer's disease (AD) onset. This neuropathological trait was recently mimicked in a novel animal model of AD, the hemizygous transgenic McGill-R-Thy1-APP (Tg(+/-)) rat. The characterization of the behavioral phenotypes in this animal model could provide a baseline of efficacy for earlier therapeutic interventions. The aim of the present study was to undertake a longitudinal study of Aß accumulation and a comprehensive behavioral evaluation of this transgenic rat model. We assessed exploratory activity, anxiety-related behaviors, recognition memory, working memory, spatial learning and reference memory at 3, 6, and 12 months of age. In parallel, we measured Aß by ELISA, Western blots and semiquantitative immunohistochemistry in hippocampal samples. SDS-soluble Aß peptide accumulated at low levels (~9 pg/mg) without differences among ages. However, Western blots showed SDS-resistant Aß oligomers (~30 kDa) at 6 and 12 months, but not at 3 months. When compared to wild-type (WT), male Tg(+/-) rats exhibited a spatial reference memory deficit in the Morris Water Maze (MWM) as early as 3 months of age, which persisted at 6 and 12 months. In addition, Tg(+/-) rats displayed a working memory impairment in the Y-maze and higher anxiety levels in the Open Field (OF) at 6 and 12 months of age, but not at 3 months. Exploratory activity in the OF was similar to that of WT at all-time points. Spatial learning in the MWM and the recognition memory, as assessed by the Novel Object Recognition Test, were unimpaired at any time point. The data from the present study demonstrate that the hemizygous transgenic McGill-R-Thy1-APP rat has a wide array of behavioral and cognitive impairments from young adulthood to middle-age. The low Aß burden and early emotional and cognitive deficits in this transgenic rat model supports its potential use for drug discovery purposes in early AD.

18.
Biosalud ; 13(1): 65-75, ene.-jun. 2014. ilus, tab
Article in Spanish | LILACS | ID: lil-734960

ABSTRACT

Se analizó la literatura científica de los últimos 10 años en las bases de datos BBCS-LILACS, Fuente Académica, IB-PsycINFO, IB-SSCI, IB-SciELO, SCOPUS y SCIRUS, estudiando las implicaciones terapéuticas de la mutación del gen ABCB1 en perros; esta mutación que consiste en la deleción de 4 pares de bases que provocan un codón de terminación prematuro, es la responsable de la ausencia de la glicoproteína P en la barrera hematoencefálica, la carencia de esta glicoproteína priva al cerebro de la protección de una bomba de eflujo frente a múltiples xenobióticos. Además, se describen los cambios farmacocinéticos y las intoxicaciones medicamentosas resultantes de esta mutación y se presenta una lista, extraída de los distintos estudios, donde se copilan algunos fármacos sustratos para la glicoproteína P y los medicamentos que pueden inhibir dicha glicoproteína, todos ellos capaces de inducir severos efectos adversos en los perros con dicha mutación.


We conducted a review of the results of the studies of the last ten years from the data bases BBCS-LILACS, IB- PsycINFO, IB-SSCI, IB.SciELO, SCOPUS and SCIRUS, about the therapeutic implications of the gene mutation ABCB1 in dogs. This mutation is responsible for the absence of the glycoprotein P in the blood-brain barrier, depriving the brain of an efflux pump to protect against various xenobiotics. Furthermore, we described the pharmacokinetic changes and drug poisoning resulting from this mutation. We also present a list of drug substrates for the glycoprotein P and the medications that can inhibit said glycoprotein, all of them capable of inducing severe side effects in dogs with the mutation.

19.
Mol Neurodegener ; 9: 5, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24405716

ABSTRACT

BACKGROUND: Familial British and Familial Danish dementias (FBD and FDD, respectively) are associated with mutations in the BRI2 gene. Processing of the mutated BRI2 protein leads to the accumulation in the brain of the 34-mer amyloid Bri (ABri) and amyloid Dan (ADan) peptides, accompanied by neurofibrillary tangles. Recently, transgenic mice successfully reproduced different aspects of FDD, while modeling of FBD in vivo has been more difficult. In this work we have modeled FBD and FDD in Drosophila and tested the hypothesis that ABri and ADan are differentially neurotoxic. RESULTS: By using site-directed insertion, we generated transgenic lines carrying ABri, ADan, Bri2-23 (the normal product of wild-type BRI2 processing) and amyloid-ß (Aß) 1-42 as a well-characterized neurotoxic peptide, alone or with a His-tag. Therefore, we avoided random insertion effects and were able to compare levels of accumulation accurately. Peptides were expressed with the GAL4-Upstream Activating Sequence (UAS) system using specific drivers. Despite low levels of expression, toxicity in the eye was characterized by mild disorganization of ommatidia and amyloid peptides accumulation. The highest toxicity was seen for ADan, followed by Aß42 and ABri. Pan-neuronal expression in the CNS revealed an age-dependent toxicity of amyloid peptides as determined by the ability of flies to climb in a geotaxis paradigm when compared to Bri2-23. This effect was stronger for ADan, detected at 7 days post-eclosion, and followed by ABri and Aß42, whose toxicity became evident after 15 and 21 days, respectively. Histological analysis showed mild vacuolization and thioflavine-S-negative deposits of amyloid peptides. In contrast, the over-expression of amyloid peptides in the specific subset of lateral neurons that control circadian locomotor activity showed no toxicity. CONCLUSIONS: Our results support the differential neurotoxicity of ADan and ABri in the Drosophila eye and CNS at low expression levels. Such differences may be partially attributed to rates of aggregation and accumulation. In the CNS, both peptides appear to be more neurotoxic than wild-type Aß42. These Drosophila models will allow a systematic and unambiguous comparison of differences and similarities in the mechanisms of toxicity of diverse amyloid peptides associated with dementia.


Subject(s)
Cataract , Cerebellar Ataxia , Cerebral Amyloid Angiopathy, Familial , Deafness , Dementia , Disease Models, Animal , Membrane Glycoproteins/toxicity , Adaptor Proteins, Signal Transducing , Amyloid Neuropathies, Familial , Animals , Animals, Genetically Modified , Cataract/genetics , Cerebellar Ataxia/genetics , Cerebral Amyloid Angiopathy, Familial/genetics , Deafness/genetics , Dementia/genetics , Drosophila melanogaster , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism
20.
Mol Cell Neurosci ; 56: 298-306, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23851186

ABSTRACT

Neuronal connectivity and synaptic remodeling are fundamental substrates for higher brain functions. Understanding their dynamics in the mammalian allocortex emerges as a critical step to tackle the cellular basis of cognitive decline that occurs during normal aging and in neurodegenerative disorders. In this work we have designed a novel approach to assess alterations in the dynamics of functional and structural connectivity elicited by chronic cell-autonomous overexpression of the human amyloid precursor protein (hAPP). We have taken advantage of the fact that the hippocampus continuously generates new dentate granule cells (GCs) to probe morphofunctional development of GCs expressing different variants of hAPP in a healthy background. hAPP was expressed together with a fluorescent reporter in neural progenitor cells of the dentate gyrus of juvenile mice by retroviral delivery. Neuronal progeny was analyzed several days post infection (dpi). Amyloidogenic cleavage products of hAPP such as the ß-C terminal fragment (ß-CTF) induced a substantial reduction in glutamatergic connectivity at 21 dpi, at which time new GCs undergo active growth and synaptogenesis. Interestingly, this effect was transient, since the strength of glutamatergic inputs was normal by 35 dpi. This delay in glutamatergic synaptogenesis was paralleled by a decrease in dendritic length with no changes in spine density, consistent with a protracted dendritic development without alterations in synapse formation. Finally, similar defects in newborn GC development were observed by overexpression of α-CTF, a non-amyloidogenic cleavage product of hAPP. These results indicate that hAPP can elicit protracted dendritic development independently of the amyloidogenic processing pathway.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Dendrites/metabolism , Hippocampus/cytology , Neurogenesis , Amyloid beta-Protein Precursor/genetics , Animals , Dendrites/physiology , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Dentate Gyrus/physiology , Excitatory Postsynaptic Potentials , Female , Hippocampus/growth & development , Hippocampus/physiology , Humans , Mice , Mice, Inbred C57BL , Nerve Net/cytology , Nerve Net/growth & development , Nerve Net/physiology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...