Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Mol Neurodegener ; 15(1): 35, 2020 06 09.
Article En | MEDLINE | ID: mdl-32517777

BACKGROUND: The apolipoprotein E (APOE) gene exists in three isoforms in humans: APOE2, APOE3 and APOE4. APOE4 causes structural and functional alterations in normal brains, and is the strongest genetic risk factor of the sporadic form of Alzheimer's disease (LOAD). Research on APOE4 has mainly focused on the neuronal damage caused by defective cholesterol transport and exacerbated amyloid-ß and Tau pathology. The impact of APOE4 on non-neuronal cell functions has been overlooked. Astrocytes, the main producers of ApoE in the healthy brain, are building blocks of neural circuits, and Ca2+ signaling is the basis of their excitability. Because APOE4 modifies membrane-lipid composition, and lipids regulate Ca2+ channels, we determined whether APOE4 dysregulates Ca2+signaling in astrocytes. METHODS: Ca2+ signals were recorded in astrocytes in hippocampal slices from APOE3 and APOE4 gene targeted replacement male and female mice using Ca2+ imaging. Mechanistic analyses were performed in immortalized astrocytes. Ca2+ fluxes were examined with pharmacological tools and Ca2+ probes. APOE3 and APOE4 expression was manipulated with GFP-APOE vectors and APOE siRNA. Lipidomics of lysosomal and whole-membranes were also performed. RESULTS: We found potentiation of ATP-elicited Ca2+responses in APOE4 versus APOE3 astrocytes in male, but not female, mice. The immortalized astrocytes modeled the male response, and showed that Ca2+ hyperactivity associated with APOE4 is caused by dysregulation of Ca2+ handling in lysosomal-enriched acidic stores, and is reversed by the expression of APOE3, but not of APOE4, pointing to loss of function due to APOE4 malfunction. Moreover, immortalized APOE4 astrocytes are refractory to control of Ca2+ fluxes by extracellular lipids, and present distinct lipid composition in lysosomal and plasma membranes. CONCLUSIONS: Immortalized APOE4 versus APOE3 astrocytes present: increased Ca2+ excitability due to lysosome dysregulation, altered membrane lipidomes and intracellular cholesterol distribution, and impaired modulation of Ca2+ responses upon changes in extracellular lipids. Ca2+ hyperactivity associated with APOE4 is found in astrocytes from male, but not female, targeted replacement mice. The study suggests that, independently of Aß and Tau pathologies, altered astrocyte excitability might contribute to neural-circuit hyperactivity depending on APOE allele, sex and lipids, and supports lysosome-targeted therapies to rescue APOE4 phenotypes in LOAD.


Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Astrocytes/metabolism , Calcium/metabolism , Lysosomes/metabolism , Alzheimer Disease/metabolism , Animals , Apolipoprotein E3/metabolism , Cholesterol/metabolism , Female , Hippocampus/metabolism , Male , Mice, Transgenic , Neurons/metabolism
2.
Glia ; 68(1): 5-26, 2020 01.
Article En | MEDLINE | ID: mdl-31058383

Systems neuroscience is still mainly a neuronal field, despite the plethora of evidence supporting the fact that astrocytes modulate local neural circuits, networks, and complex behaviors. In this article, we sought to identify which types of studies are necessary to establish whether astrocytes, beyond their well-documented homeostatic and metabolic functions, perform computations implementing mathematical algorithms that sub-serve coding and higher-brain functions. First, we reviewed Systems-like studies that include astrocytes in order to identify computational operations that these cells may perform, using Ca2+ transients as their encoding language. The analysis suggests that astrocytes may carry out canonical computations in a time scale of subseconds to seconds in sensory processing, neuromodulation, brain state, memory formation, fear, and complex homeostatic reflexes. Next, we propose a list of actions to gain insight into the outstanding question of which variables are encoded by such computations. The application of statistical analyses based on machine learning, such as dimensionality reduction and decoding in the context of complex behaviors, combined with connectomics of astrocyte-neuronal circuits, is, in our view, fundamental undertakings. We also discuss technical and analytical approaches to study neuronal and astrocytic populations simultaneously, and the inclusion of astrocytes in advanced modeling of neural circuits, as well as in theories currently under exploration such as predictive coding and energy-efficient coding. Clarifying the relationship between astrocytic Ca2+ and brain coding may represent a leap forward toward novel approaches in the study of astrocytes in health and disease.


Astrocytes/physiology , Brain/physiology , Neurosciences/methods , Systems Biology/methods , Animals , Astrocytes/chemistry , Brain/cytology , Brain Chemistry/physiology , Humans , Neurons/chemistry , Neurons/physiology , Neurosciences/trends , Optogenetics/methods , Systems Biology/trends
3.
J Am Chem Soc ; 141(18): 7628-7636, 2019 05 08.
Article En | MEDLINE | ID: mdl-31010281

Light-triggered reversible modulation of physiological functions offers the promise of enabling on-demand spatiotemporally controlled therapeutic interventions. Optogenetics has been successfully implemented in the heart, but significant barriers to its use in the clinic remain, such as the need for genetic transfection. Herein, we present a method to modulate cardiac function with light through a photoswitchable compound and without genetic manipulation. The molecule, named PAI, was designed by introduction of a photoswitch into the molecular structure of an M2 mAChR agonist. In vitro assays revealed that PAI enables light-dependent activation of M2 mAChRs. To validate the method, we show that PAI photoisomers display different cardiac effects in a mammalian animal model, and demonstrate reversible, real-time photocontrol of cardiac function in translucent wildtype tadpoles. PAI can also effectively activate M2 receptors using two-photon excitation with near-infrared light, which overcomes the scattering and low penetration of short-wavelength illumination, and offers new opportunities for intravital imaging and control of cardiac function.


Muscarinic Agonists/pharmacology , Parasympathetic Nervous System/drug effects , Receptor, Muscarinic M2/agonists , Sympathetic Nervous System/drug effects , Animals , Dose-Response Relationship, Drug , Heart Rate/drug effects , Infrared Rays , Molecular Docking Simulation , Molecular Structure , Muscarinic Agonists/chemical synthesis , Muscarinic Agonists/chemistry , Photochemical Processes , Rats , Rats, Wistar , Stereoisomerism , Structure-Activity Relationship , Xenopus
4.
Glia ; 66(8): 1724-1735, 2018 08.
Article En | MEDLINE | ID: mdl-29575211

The prevalent view in neuroenergetics is that glucose is the main brain fuel, with neurons being mostly oxidative and astrocytes glycolytic. Evidence supporting that astrocyte mitochondria are functional has been overlooked. Here we sought to determine what is unique about astrocyte mitochondria by performing unbiased statistical comparisons of the mitochondriome in astrocytes and neurons. Using MitoCarta, a compendium of mitochondrial proteins, together with transcriptomes of mouse neurons and astrocytes, we generated cell-specific databases of nuclear genes encoding for mitochondrion proteins, ranked according to relative expression. Standard and in-house Gene Set Enrichment Analyses (GSEA) of five mouse transcriptomes revealed that genes encoding for enzymes involved in fatty acid oxidation (FAO) and amino acid catabolism are consistently more expressed in astrocytes than in neurons. FAO and oxidative-metabolism-related genes are also up-regulated in human cortical astrocytes versus the whole cortex, and in adult astrocytes versus fetal astrocytes. We thus present the first evidence of FAO in human astrocytes. Further, as shown in vitro, FAO coexists with glycolysis in astrocytes and is inhibited by glutamate. Altogether, these analyses provide arguments against the glucose-centered view of energy metabolism in astrocytes and reveal mitochondria as specialized organelles in these cells.


Astrocytes/metabolism , Energy Metabolism/physiology , Fatty Acids/metabolism , Glycolysis/physiology , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Animals , Glutamic Acid/metabolism , Humans , Lipid Metabolism , Mice , Mitochondrial Proteins/metabolism , Neurons/metabolism , Oxidation-Reduction
6.
Sci Rep ; 7(1): 6390, 2017 07 25.
Article En | MEDLINE | ID: mdl-28743894

The cyclic AMP response element binding protein (CREB) is a primary hub of activity-driven genetic programs in neurons controlling plasticity, neurogenesis and survival. By contrast, the gene networks coordinated by CREB in astrocytes are unknown despite the fact that the astrocytic CREB is also activity-driven and neuroprotective. Herein we identified the transcriptional programs regulated by CREB in astrocytes as compared to neurons using, as study materials, transcriptome databases of astrocyte exposed to well-known activators of CREB-dependent transcription as well as publicly available transcriptomes of neuronal cultures. Functional CREB signatures were extracted from the transcriptomes using Gene Ontology, adult-brain gene lists generated by Translating Ribosome Affinity Purification (TRAP) and CREB-target gene repositories. We found minimal overlap between CREB signatures in astrocytes and neurons. In astrocytes, the top triad of functions regulated by CREB consists of 'Gene expression', 'Mitochondria', and 'Signalling', while in neurons it is 'Neurotransmission', 'Signalling' and 'Gene expression', the latter two being represented by different genes from those in astrocytes. The newly generated databases will provide a tool to explore novel means whereby CREB impinges on brain functions requiring adaptive, long-lasting changes by coordinating transcriptional cascades in astrocytes.


Astrocytes/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Profiling/methods , Gene Regulatory Networks , Neurons/metabolism , Animals , Astrocytes/cytology , Cells, Cultured , Databases, Genetic , Gene Expression Regulation , Neurons/cytology , Oligonucleotide Array Sequence Analysis , Organ Specificity , Rats , Rats, Sprague-Dawley , Transcription, Genetic
7.
Nat Commun ; 8: 15132, 2017 05 02.
Article En | MEDLINE | ID: mdl-28462931

The influence that neurons exert on astrocytic function is poorly understood. To investigate this, we first developed a system combining cortical neurons and astrocytes from closely related species, followed by RNA-seq and in silico species separation. This approach uncovers a wide programme of neuron-induced astrocytic gene expression, involving Notch signalling, which drives and maintains astrocytic maturity and neurotransmitter uptake function, is conserved in human development, and is disrupted by neurodegeneration. Separately, hundreds of astrocytic genes are acutely regulated by synaptic activity via mechanisms involving cAMP/PKA-dependent CREB activation. This includes the coordinated activity-dependent upregulation of major astrocytic components of the astrocyte-neuron lactate shuttle, leading to a CREB-dependent increase in astrocytic glucose metabolism and elevated lactate export. Moreover, the groups of astrocytic genes induced by neurons or neuronal activity both show age-dependent decline in humans. Thus, neurons and neuronal activity regulate the astrocytic transcriptome with the potential to shape astrocyte-neuron metabolic cooperation.


Astrocytes/metabolism , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental , Neurons/metabolism , Tauopathies/genetics , Animals , Astrocytes/cytology , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cell Communication , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Coculture Techniques , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Embryo, Mammalian , Gene Expression Profiling , Glucose/metabolism , High-Throughput Nucleotide Sequencing , Humans , Lactic Acid/metabolism , Membrane Potentials/physiology , Mice, Knockout , Neurons/cytology , Rats, Sprague-Dawley , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Tauopathies/metabolism , Tauopathies/pathology
8.
Trends Mol Med ; 23(6): 486-500, 2017 06.
Article En | MEDLINE | ID: mdl-28499701

Central nervous system (CNS) therapeutics based on the theoretical framework of neuroinflammation have only barely succeeded. We argue that a problem may be the wrong use of the term 'neuroinflammation' as a distinct nosological entity when, based on recent evidence, it may not explain CNS disease pathology. Indeed, the terms 'neuroinflammation' and 'glia' could be obsolete. First, unbiased molecular profiling of CNS cell populations and individual cells reveals striking phenotypic heterogeneity in health and disease. Second, astrocytes, microglia, oligodendrocytes, and NG2 cells may contribute to higher-brain functions by performing actions beyond housekeeping. We propose that CNS diseases be viewed as failed circuits caused in part by disease-specific dysfunction of cells traditionally called 'glia', and hence, favor therapies promoting their functional recovery.


Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Neuroglia , Animals , Central Nervous System Diseases/therapy , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Neuroglia/metabolism , Neuroglia/pathology
9.
Glia ; 64(5): 853-74, 2016 May.
Article En | MEDLINE | ID: mdl-26880229

The clinical challenge in acute injury as in traumatic brain injury (TBI) is to halt the delayed neuronal loss that occurs hours and days after the insult. Here we report that the activation of CREB-dependent transcription in reactive astrocytes prevents secondary injury in cerebral cortex after experimental TBI. The study was performed in a novel bitransgenic mouse in which a constitutively active CREB, VP16-CREB, was targeted to astrocytes with the Tet-Off system. Using histochemistry, qPCR, and gene profiling we found less neuronal death and damage, reduced macrophage infiltration, preserved mitochondria, and rescued expression of genes related to mitochondrial metabolism in bitransgenic mice as compared to wild type littermates. Finally, with meta-analyses using publicly available databases we identified a core set of VP16-CREB candidate target genes that may account for the neuroprotective effect. Enhancing CREB activity in astrocytes thus emerges as a novel avenue in acute brain post-injury therapeutics.


Astrocytes/metabolism , Brain Injuries/pathology , Brain Injuries/therapy , CREB-Binding Protein/metabolism , Animals , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Astrocytes/drug effects , CREB-Binding Protein/genetics , Cells, Cultured , Disease Models, Animal , Etoposide/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Inflammation/etiology , Inflammation/prevention & control , Male , Meta-Analysis as Topic , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neurofilament Proteins/metabolism
10.
Neurobiol Aging ; 34(12): 2853-65, 2013 Dec.
Article En | MEDLINE | ID: mdl-23891486

α-Synuclein (AS) plays a crucial role in Parkinson's disease pathogenesis. AS is normally secreted from neuronal cells and can thus exert paracrine effects. We have previously demonstrated that naturally secreted AS species, derived from SH-SY5Y cells inducibly overexpressing human wild type AS, can be toxic to recipient neuronal cells. In the current study, we show that application of secreted AS alters membrane fluidity and increases calcium (Ca2+) entry. This influx is reduced on pharmacological inhibition of voltage-operated Ca2+ channels. Although no change in free cytosolic Ca2+ levels is observed, a significantly increased mitochondrial Ca2+ sequestration is found in recipient cells. Application of voltage-operated Ca2+ channel blockers or Ca2+ chelators abolishes AS-mediated toxicity. AS-treated cells exhibit increased calpain activation, and calpain inhibition greatly alleviates the observed toxicity. Collectively, our data suggest that secreted AS exerts toxicity through engagement, at least in part, of the Ca2+ homeostatic machinery. Therefore, manipulating Ca2+ signaling pathways might represent a potential therapeutic strategy for Parkinson's disease.


Calcium/metabolism , Calcium/physiology , Homeostasis/drug effects , Neurons/drug effects , Neurons/metabolism , Parkinson Disease/genetics , alpha-Synuclein/toxicity , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Humans , Membrane Fluidity/drug effects , Molecular Targeted Therapy , Parkinson Disease/drug therapy , alpha-Synuclein/metabolism
11.
Biochem Biophys Res Commun ; 427(2): 326-9, 2012 Oct 19.
Article En | MEDLINE | ID: mdl-22995315

Evidence suggests that ß-Adrenergic receptor signaling increases heart rate and force through not just cyclic AMP but also the Ca(2+)-releasing second messengers NAADP (nicotinic acid adenine dinucleotide phosphate) and cADPR (cyclic ADP-ribose). Nevertheless, proof of the physiological relevance of these messengers requires direct measurements of their levels in response to receptor stimulation. Here we report that in intact Langendorff-perfused hearts ß-adrenergic stimulation increased both messengers, with NAADP being transient and cADPR being sustained. Both NAADP and cADPR have physiological and therefore pathological relevance by providing alternative drug targets in the ß-adrenergic receptor signaling pathway.


Cyclic ADP-Ribose/metabolism , Myocardium/metabolism , NADP/analogs & derivatives , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Guinea Pigs , Heart/drug effects , In Vitro Techniques , NADP/metabolism , Signal Transduction
12.
Glia ; 60(9): 1330-44, 2012 Sep.
Article En | MEDLINE | ID: mdl-22593004

In neurons, it is well established that CREB contributes to learning and memory by orchestrating the translation of experience into the activity-dependent (i.e., driven by neurotransmitters) transcription of plasticity-related genes. The activity-dependent CREB-triggered transcription requires the concerted action of cyclic AMP/protein kinase A and Ca(2+) /calcineurin via the CREB-regulated transcription co-activator (CRTC). It is not known, however, whether a comparable molecular sequence occurs in astrocytes, despite the unquestionable contribution of these cells to brain plasticity. Here we sought to determine whether and how ATP and noradrenaline cause CREB-dependent transcription in rat cortical astrocyte cultures. Both transmitters induced CREB phosphorylation (Western Blots), CREB-dependent transcription (CRE-luciferase reporter assays), and the transcription of Bdnf, a canonical regulator of synaptic plasticity (quantitative RT-PCR). We indentified a Ca(2+) and diacylglycerol-independent protein kinase C at the uppermost position of the cascade leading to CREB-dependent transcription. Notably, CREB-dependent transcription was partially dependent on ERK1/2 and CRTC, but independent of cyclic AMP/protein kinase A or Ca(2+) /calcineurin. We conclude that ATP and noradrenaline activate CREB-dependent transcription in cortical astrocytes via an atypical protein kinase C. It is of relevance that the signaling involved be starkly different to the one described in neurons since there is no convergence of Ca(2+) and cyclic AMP-dependent pathways on CRTC, which, moreover, exerts a modulatory rather than a central role. Our data thus point to the existence of an alternative, non-neuronal, glia-based role of CREB in plasticity.


Adenosine Triphosphate/pharmacology , Astrocytes/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Norepinephrine/pharmacology , Signal Transduction/drug effects , Animals , Astrocytes/cytology , Astrocytes/metabolism , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Glutamic Acid/pharmacology , HEK293 Cells , Humans , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Transcription, Genetic/drug effects
13.
Proteomics ; 12(1): 145-56, 2012 Jan.
Article En | MEDLINE | ID: mdl-22065602

Glial cells in the peripheral nervous system, such as Schwann cells, respond to nucleotides, which play an important role in axonal regeneration and myelination. Metabotropic P2Y receptor agonists are promising therapeutic molecules for peripheral neuropathies. Nevertheless, the proteomic mechanisms involved in nucleotide action on Schwann cells remain unknown. Here, we studied intracellular protein changes in RT4-D6P2T Schwann cells after treatment with nucleotides and Nucleo CMP Forte (CMPF), a nucleotide-based drug. After treatment with CMPF, 2-D DIGE revealed 11 differential gel spots, which were all upregulated. Among these, six different proteins were identified by MS. Some of these proteins are involved in actin remodelling (actin-related protein, Arp3), membrane vesicle transport (Rab GDP dissociation inhibitor ß, Rab GDI), and the endoplasmic reticulum stress response (protein disulfide isomerase A3, PDI), which are hallmarks of a possible P2Y receptor signalling pathway. Expression of P2Y receptors in RT4-D6P2T cells was demonstrated by RT-PCR and a transient elevation of intracellular calcium measured in response to UTP. Actin reorganisation was visualized after UTP treatment using phalloidin-FITC staining and was blocked by the P2Y antagonist suramin, which also inhibited Arp3, Rab GDI, and PDI protein upregulation. Our data indicate that extracellular UTP interacts with Schwann P2Y receptors and activates molecular machinery that induces changes in the glial cell cytoskeleton.


Proteome/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y/metabolism , Uridine Triphosphate/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cytidine Monophosphate/pharmacology , Cytoskeletal Proteins/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Gene Expression , Neurilemmoma , Proteome/genetics , Rats , Receptors, Purinergic P2Y/genetics , Two-Dimensional Difference Gel Electrophoresis , Up-Regulation/drug effects
14.
FEBS Lett ; 585(14): 2300-6, 2011 Jul 21.
Article En | MEDLINE | ID: mdl-21664355

Intracellular Ca(2+) signals provide astrocytes with a specific form of excitability that enables them to regulate synaptic transmission. In this study, we demonstrate that NAADP-AM, a membrane-permeant analogue of the new second messenger nicotinic acid-adenine dinucleotide phosphate (NAADP), mobilizes Ca(2+) in astrocytes and that the response is blocked by Ned-19, an antagonist of NAADP signalling. We also show that NAADP receptors are expressed in lysosome-related acidic vesicles. Pharmacological disruption of either NAADP or lysosomal signalling reduced Ca(2+) responses induced by ATP and endothelin-1, but not by bradykinin. Furthermore, ATP increased endogenous NAADP levels. Overall, our data provide evidence for NAADP being an intracellular messenger for agonist-mediated calcium signalling in astrocytes.


Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Calcium Signaling/physiology , Calcium/metabolism , NADP/analogs & derivatives , Animals , Astrocytes/cytology , Bradykinin/metabolism , Cells, Cultured , Chelating Agents/metabolism , Egtazic Acid/metabolism , Endothelin-1/metabolism , NADP/metabolism , Rats , Rats, Sprague-Dawley
15.
Front Aging Neurosci ; 2: 142, 2010.
Article En | MEDLINE | ID: mdl-21152343

The use of non-steroidal anti-inflammatory drugs (NSAIDs) in Alzheimer's disease (AD) is controversial because conclusions from numerous epidemiological studies reporting delayed onset of AD in NSAID users have not been corroborated in clinical trials. The purpose of this personal view is to revise the case for NSAIDs in AD therapeutics in light of: (i) the last report from the only primary prevention trial in AD, ADAPT, which, although incomplete, points to significant protection in long-term naproxen users, and (ii) the recently proposed dynamic model of AD evolution. The model contends that there is a clinical silent phase in AD that can last up to 20 years, the duration depending on life style habits, genetic factors, or cognitive reserve. The failure of many purported disease-modifying drugs in AD clinical trials is forcing the view that treatments will only be efficacious if administered pre-clinically. Here we will argue that NSAIDs failed in clinical trials because they are disease-modifying drugs, and they should be administered in early stages of the disease. A complete prevention trial in cognitively normal individuals is thus called for. Further, the shift of anti-inflammatory treatment to early stages uncovers a knowledge void about the targets of NSAIDs in asymptomatic individuals. AD researchers have mostly relied on post-mortem analysis of Aß plaque-laden brains from demented patients or animal models, thus drawing conclusions about AD pathogenesis based on late symptoms. We will discuss evidence in support that defective, not excessive, inflammation underlies AD pathogenesis, that NSAIDs are multifunctional drugs acting on inflammatory and non-inflammatory targets, and that astrocytes and microglia may play differing roles in disease progression, with an emphasis of ApoEε4 as a key, undervalued target of NSAIDs. According to a meta-analysis of epidemiological data, NSAIDs afford an average protection of 58%. If this figure is true, and translated into patient numbers, NSAID treatment may revive as a worth pursuing strategy to significantly reduce the socio-economical burden imposed by AD.

16.
J Neurosci ; 30(28): 9402-10, 2010 Jul 14.
Article En | MEDLINE | ID: mdl-20631169

Activity-dependent gene expression mediating changes of synaptic efficacy is important for memory storage, but the mechanisms underlying gene transcriptional changes in age-related memory disorders are poorly understood. In this study, we report that gene transcription mediated by the cAMP-response element binding protein (CREB)-regulated transcription coactivator CRTC1 is impaired in neurons and brain from an Alzheimer's disease (AD) transgenic mouse expressing the human beta-amyloid precursor protein (APP(Sw,Ind)). Suppression of CRTC1-dependent gene transcription by beta-amyloid (Abeta) in response to cAMP and Ca(2+) signals is mediated by reduced calcium influx and disruption of PP2B/calcineurin-dependent CRTC1 dephosphorylation at Ser151. Consistently, expression of CRTC1 or active CRTC1 S151A and calcineurin mutants reverse the deficits on CRTC1 transcriptional activity in APP(Sw,Ind) neurons. Inhibition of calcium influx by pharmacological blockade of L-type voltage-gated calcium channels (VGCCs), but not by blocking NMDA or AMPA receptors, mimics the decrease on CRTC1 transcriptional activity observed in APP(Sw,Ind) neurons, whereas agonists of L-type VGCCs reverse efficiently these deficits. Consistent with a role of CRTC1 on Abeta-induced synaptic and memory dysfunction, we demonstrate a selective reduction of CRTC1-dependent genes related to memory (Bdnf, c-fos, and Nr4a2) coinciding with hippocampal-dependent spatial memory deficits in APP(Sw,Ind) mice. These findings suggest that CRTC1 plays a key role in coupling synaptic activity to gene transcription required for hippocampal-dependent memory, and that Abeta could disrupt cognition by affecting CRTC1 function.


Amyloid beta-Protein Precursor/genetics , Cerebral Cortex/metabolism , Memory/physiology , Neurons/metabolism , Transcription Factors/genetics , Transcription, Genetic/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Behavior, Animal/physiology , Blotting, Western , Calcineurin/metabolism , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Chromatin Immunoprecipitation , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Transgenic , Neurons/cytology , Phosphorylation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
17.
J Biol Chem ; 284(47): 32359-69, 2009 Nov 20.
Article En | MEDLINE | ID: mdl-19778898

The biogenesis of lipid droplets (LD) induced by serum depends on group IVA phospholipase A(2) (cPLA(2)alpha). This work dissects the pathway leading to cPLA(2)alpha activation and LD biogenesis. Both processes were Ca(2+)-independent, as they took place after pharmacological blockade of Ca(2+) transients elicited by serum or chelation with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester). The single mutation D43N in cPLA(2)alpha, which abrogates its Ca(2+) binding capacity and translocation to membranes, did not affect enzyme activation and formation of LD. In contrast, the mutation S505A did not affect membrane relocation of the enzyme in response to Ca(2+) but prevented its phosphorylation, activation, and the appearance of LD. Expression of specific activators of different mitogen-activated protein kinases showed that phosphorylation of cPLA(2)alpha at Ser-505 is due to JNK. This was confirmed by pharmacological inhibition and expression of a dominant-negative form of the upstream activator MEKK1. LD biogenesis was accompanied by increased synthesis of ceramide 1-phosphate. Overexpression of its synthesizing enzyme ceramide kinase increased phosphorylation of cPLA(2)alpha at Ser-505 and formation of LD, and its down-regulation blocked the phosphorylation of cPLA(2)alpha and LD biogenesis. These results demonstrate that LD biogenesis induced by serum is regulated by JNK and ceramide kinase.


Group IV Phospholipases A2/metabolism , Lipids/chemistry , MAP Kinase Kinase 4/metabolism , Animals , CHO Cells , Calcium/chemistry , Calcium/metabolism , Cricetinae , Cricetulus , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, Dominant , Humans , Models, Biological , Mutation , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism
18.
J Biol Chem ; 284(9): 5697-708, 2009 Feb 27.
Article En | MEDLINE | ID: mdl-19117952

This work investigates the metabolic origin of triacylglycerol (TAG) formed during lipid droplet (LD) biogenesis induced by stress. Cytotoxic inhibitors of fatty acid synthase induced TAG synthesis and LD biogenesis in CHO-K1 cells, in the absence of external sources of fatty acids. TAG synthesis was required for LD biogenesis and was sensitive to inhibition and down-regulation of the expression of group VIA phospholipase A(2) (iPLA(2)-VIA). Induction of stress with acidic pH, C(2)-ceramide, tunicamycin, or deprivation of glucose also stimulated TAG synthesis and LD formation in a manner dependent on iPLA(2)-VIA. Overexpression of the enzyme enhanced TAG synthesis from endogenous fatty acids and LD occurrence. During stress, LD biogenesis but not TAG synthesis required phosphorylation and activation of group IVA PLA(2) (cPLA(2)alpha). The results demonstrate that iPLA(2)-VIA provides fatty acids for TAG synthesis while cPLA(2)alpha allows LD biogenesis. LD biogenesis during stress may be a survival strategy, recycling structural phospholipids into energy-generating substrates.


Group VI Phospholipases A2/metabolism , Lipid Metabolism/physiology , Oxidative Stress , Triglycerides/biosynthesis , Animals , Anti-Bacterial Agents/pharmacology , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/metabolism , Fatty Acids , Flow Cytometry , Fluorescence , Glucose/deficiency , Group VI Phospholipases A2/antagonists & inhibitors , Phosphorylation , RNA, Small Interfering/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Tunicamycin/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/genetics , Type C Phospholipases/metabolism
19.
Neuropharmacology ; 56(3): 598-609, 2009 Mar.
Article En | MEDLINE | ID: mdl-19061903

In neurogenesis, little is known about signal transduction pathways upstream of gene expression however, mounting evidence suggests that calcium release from internal stores plays a critical role. We have previously demonstrated that BM88 is a neuronal lineage-specific regulator of cell cycle exit and differentiation; we now report a link between BM88 and calcium signaling. Calcium imaging experiments revealed that P2Y-induced calcium mobilization is diminished in mouse neuroblastoma Neuro 2a cells stably transfected with BM88 (N2A-BM88 cells) as compared with N2A cells or N2A cells differentiated with retinoic acid. This effect is not restricted to N2A cells but is also observed in HeLa cells that are transiently transfected with BM88, indicating that cells of both neural and non-neural origin respond similarly. Further, activation of P2Y1 but not purinergic P2X receptors induces proliferation of N2A and to a lesser extent of N2A-BM88 cells. Conversely, knockdown of BM88 facilitates N2A cell proliferation both under stimulating and non-stimulating conditions. Importantly, N2A-BM88 cells are less susceptible to apoptosis triggered by C2-ceramide and exhibit reduced C2-ceramide-induced intracellular calcium release. Higher calcium uptake from mitochondria and/or lower calcium levels inside the endoplasmic reticulum may explain the reduced calcium mobilization in response to BM88. Overall, our data reveal a novel signaling mechanism by which BM88 interferes with calcium release from inositol 1,4,5-trisphosphate-sensitive stores and exerts anti-proliferative and anti-apoptotic functions.


Calcium Signaling/physiology , Calcium/metabolism , Intracellular Fluid/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Differentiation , Cell Proliferation , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neurogenesis , Receptors, Purinergic P2/physiology , Sequence Alignment , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Transfection , Tumor Cells, Cultured
20.
J Biol Chem ; 283(41): 27369-27382, 2008 Oct 10.
Article En | MEDLINE | ID: mdl-18632668

Lipid droplets (LD) are organelles present in all cell types, consisting of a hydrophobic core of triacylglycerols and cholesteryl esters, surrounded by a monolayer of phospholipids and cholesterol. This work shows that LD biogenesis induced by serum, by long-chain fatty acids, or the combination of both in CHO-K1 cells was prevented by phospholipase A(2) inhibitors with a pharmacological profile consistent with the implication of group IVA cytosolic phospholipase A(2) (cPLA(2)alpha). Knocking down cPLA(2)alpha expression with short interfering RNA was similar to pharmacological inhibition in terms of enzyme activity and LD biogenesis. A Chinese hamster ovary cell clone stably expressing an enhanced green fluorescent protein-cPLA(2)alpha fusion protein (EGFP-cPLA(2)) displayed higher LD occurrence under basal conditions and upon LD induction. Induction of LD took place with concurrent phosphorylation of cPLA(2)alpha at Ser(505). Transfection of a S505A mutant cPLA(2)alpha showed that phosphorylation at Ser(505) is key for enzyme activity and LD formation. cPLA(2)alpha contribution to LD biogenesis was not because of the generation of arachidonic acid, nor was it related to neutral lipid synthesis. cPLA(2)alpha inhibition in cells induced to form LD resulted in the appearance of tubulo-vesicular profiles of the smooth endoplasmic reticulum, compatible with a role of cPLA(2)alpha in the formation of nascent LD from the endoplasmic reticulum.


Endoplasmic Reticulum/enzymology , Gene Expression Regulation, Enzymologic/physiology , Group IV Phospholipases A2/metabolism , Lipid Metabolism/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Group IV Phospholipases A2/antagonists & inhibitors , Humans , Lipid Metabolism/drug effects , Phosphorylation/drug effects , RNA, Small Interfering/pharmacology , Rats
...