Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
2.
Cell Rep ; 40(10): 111271, 2022 09 06.
Article En | MEDLINE | ID: mdl-36070700

Healthy aging is an ambitious aspiration for humans, but neurodegenerative disorders, such as Alzheimer's disease (AD), strongly affect quality of life. Using an integrated omics approach, we investigate alterations in the molecular composition of postmortem hippocampus samples of healthy persons and individuals with AD. Profound differences are apparent between control and AD male and female cohorts in terms of up- and downregulated metabolic pathways. A decrease in the insulin response is evident in AD when comparing the female with the male group. The serine metabolism (linked to the glycolytic pathway and generating the N-methyl-D-aspartate [NMDA] receptor coagonist D-serine) is also significantly modulated: the D-Ser/total serine ratio represents a way to counteract age-related cognitive decline in healthy men and during AD onset in women. These results show how AD changes and, in certain respects, almost reverses sex-specific proteomic and metabolomic profiles, highlighting how different pathophysiological mechanisms are active in men and women.


Alzheimer Disease , Alzheimer Disease/metabolism , Female , Hippocampus/metabolism , Humans , Insulin/metabolism , Male , Proteomics , Quality of Life , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/metabolism
3.
Adv Exp Med Biol ; 1331: 145-165, 2021.
Article En | MEDLINE | ID: mdl-34453297

Alzheimer's disease (AD), one of the most common causes of dementia in elderly people, is characterized by progressive impairment in cognitive function, early degeneration of basal forebrain cholinergic neurons (BFCNs), abnormal metabolism of the amyloid precursor protein (APP), amyloid beta-peptide (Aß) depositions, and neurofibrillary tangles. According to the cholinergic hypothesis, dysfunction of acetylcholine-containing neurons in the basal forebrain contributes markedly to the cognitive decline observed in AD. In addition, the neurotrophic factor hypothesis posits that the loss nerve growth factor (NGF) signalling in AD may account for the vulnerability to atrophy of BFCNs and consequent impairment of cholinergic functions. Though acetylcholinesterase inhibitors provide only partial and symptomatic relief to AD patients, emerging data from in vivo magnetic resonance imaging (MRI) and positron emission tomography (PET) studies in mild cognitive impairment (MCI) and AD patients highlight the early involvement of BFCNs in MCI and the early phase of AD. These data support the cholinergic and neurotrophic hypotheses of AD and suggest new targets for AD therapy.Different mechanisms account for selective vulnerability of BFCNs to AD pathology, with regard to altered metabolism of APP and tau. In this review, we provide a general overview of the current knowledge of NGF and APP interplay, focusing on the role of APP in regulating NGF receptors trafficking/signalling and on the involvement of NGF in modulating phosphorylation of APP, which in turn controls APP intracellular trafficking and processing. Moreover, we highlight the consequences of APP interaction with p75NTR and TrkA receptor, which share the same binding site within the APP juxta-membrane domain. We underline the importance of insulin dysmetabolism in AD pathology, in the light of our recent data showing that overlapping intracellular signalling pathways stimulated by NGF or insulin can be compensatory. In particular, NGF-based signalling is able to ameliorates deficiencies in insulin signalling in the medial septum of 3×Tg-AD mice. Finally, we present an overview of NGF-regulated microRNAs (miRNAs). These small non-coding RNAs are involved in post-transcriptional regulation of gene expression , and we focus on a subset that are specifically deregulated in AD and thus potentially contribute to its pathology.


Alzheimer Disease , Amyloid beta-Protein Precursor , Aged , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Animals , Humans , Mice , Nerve Growth Factor , Neurons
4.
Cells ; 9(8)2020 07 30.
Article En | MEDLINE | ID: mdl-32751526

Alzheimer's disease (AD) is an incurable neurodegenerative disorder with a few early detection strategies. We previously proposed the amyloid precursor protein (APP) tyrosine 682 (Tyr682) residue as a valuable target for the development of new innovative pharmacologic or diagnostic interventions in AD. Indeed, when APP is phosphorylated at Tyr682, it is forced into acidic neuronal compartments where it is processed to generate neurotoxic amyloid ß peptides. Of interest, Fyn tyrosine kinase (TK) interaction with APP Tyr682 residue increases in AD neurons. Here we proved that when Fyn TK was overexpressed it elicited APP Tyr682 phosphorylation in neurons from healthy donors and promoted the amyloidogenic APP processing with Aß peptides accumulation and neuronal death. Phosphorylation of APP at Tyr (pAPP-Tyr) increased in neurons of AD patients and AD neurons that exhibited high pAPP-Tyr also had higher Fyn TK activity. Fyn TK inhibition abolished the pAPP-Tyr and reduced Aß42 secretion in AD neurons. In addition, the multidomain adaptor protein Fe65 controlled the Fyn-mediated pAPP-Tyr, warranting the possibility of targeting the Fe65-APP-Fyn pathway to develop innovative strategies in AD. Altogether, these results strongly emphasize the relevance of focusing on pAPP Tyr682 either for diagnostic purposes, as an early biomarker of the disease, or for pharmacological targeting, using Fyn TKI.


Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Tyrosine/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Biomarkers/metabolism , Cells, Cultured , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Middle Aged , Neural Stem Cells/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/genetics , Transfection
5.
Biochim Biophys Acta Proteins Proteom ; 1868(10): 140471, 2020 10.
Article En | MEDLINE | ID: mdl-32561430

Free d-aspartate is abundant in the mammalian embryonic brain. However, following the postnatal onset of the catabolic d-aspartate oxidase (DDO) activity, cerebral d-aspartate levels drastically decrease, remaining constantly low throughout life. d-Aspartate stimulates both glutamatergic NMDA receptors (NMDARs) and metabotropic Glu5 receptors. In rodents, short-term d-aspartate exposure increases spine density and synaptic plasticity, and improves cognition. Conversely, persistently high d-Asp levels produce NMDAR-dependent neurotoxic effects, leading to precocious neuroinflammation and cell death. These pieces of evidence highlight the dichotomous impact of d-aspartate signaling on NMDAR-dependent processes and, in turn, unveil a neuroprotective role for DDO in preventing the detrimental effects of excessive d-aspartate stimulation during aging. Here, we will focus on the in vivo influence of altered d-aspartate metabolism on the modulation of glutamatergic functions and its involvement in translational studies. Finally, preliminary data on the role of embryonic d-aspartate in the mouse brain will also be reviewed.


Brain/metabolism , D-Aspartic Acid/metabolism , Mammals/metabolism , Neurogenesis , Age Factors , Aging/genetics , Aging/metabolism , Animals , Biomarkers , Brain/anatomy & histology , Brain/growth & development , D-Aspartate Oxidase/genetics , D-Aspartate Oxidase/metabolism , Dietary Supplements , Disease Susceptibility , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Memory , Neuroprotection , Pregnancy , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
6.
Exp Neurol ; 317: 51-65, 2019 07.
Article En | MEDLINE | ID: mdl-30822420

In mammals, free d-aspartate (D-Asp) is abundant in the embryonic brain, while levels remain very low during adulthood as a result of the postnatal expression and activity of the catabolizing enzyme d-aspartate oxidase (DDO). Previous studies have shown that long-lasting exposure to nonphysiological, higher D-Asp concentrations in Ddo knockout (Ddo-/-) mice elicits a precocious decay of synaptic plasticity and cognitive functions, along with a dramatic age-dependent expression of active caspase 3, associated with increased cell death in different brain regions, including hippocampus, prefrontal cortex, and substantia nigra pars compacta. Here, we investigate the yet unclear molecular and cellular events associated with the exposure of abnormally high D-Asp concentrations in cortical primary neurons and in the brain of Ddo-/- mice. For the first time, our in vitro findings document that D-Asp induces in a time-, dose-, and NMDA receptor-dependent manner alterations in JNK and Tau phosphorylation levels, associated with pronounced cell death in primary cortical neurons. Moreover, observations obtained in Ddo-/- animals confirmed that high in vivo levels of D-Asp altered cortical JNK signaling, Tau phosphorylation and enhanced protein SUMOylation, indicating a robust indirect role of DDO activity in regulating these biochemical NMDA receptor-related processes. Finally, no gross modifications in D-Asp concentrations and DDO mRNA expression were detected in the cortex of patients with Alzheimer's disease when compared to age-matched healthy controls.


Alzheimer Disease/pathology , Cell Death/drug effects , Cerebral Cortex/pathology , D-Aspartate Oxidase/metabolism , D-Aspartic Acid/pharmacology , MAP Kinase Kinase 4/metabolism , Neurons/pathology , Receptors, N-Methyl-D-Aspartate/drug effects , Sumoylation/drug effects , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Animals , Cognition Disorders/psychology , D-Aspartate Oxidase/genetics , Female , Humans , Mice , Mice, Knockout , Middle Aged , Neuronal Plasticity/drug effects , Neurons/drug effects , Phosphorylation/drug effects , Pregnancy , Primary Cell Culture
8.
Mol Neurobiol ; 56(1): 535-552, 2019 Jan.
Article En | MEDLINE | ID: mdl-29736736

Basal forebrain cholinergic neurons (BFCN) are key modulators of learning and memory and are high energy-demanding neurons. Impaired neuronal metabolism and reduced insulin signaling, known as insulin resistance, has been reported in the early phase of Alzheimer's disease (AD), which has been suggested to be "Type 3 Diabetes." We hypothesized that BFCN may develop insulin resistance and their consequent failure represents one of the earliest event in AD. We found that a condition reminiscent of insulin resistance occurs in the medial septum of 3 months old 3×Tg-AD mice, reported to develop typical AD histopathology and cognitive deficits in adulthood. Further, we obtained insulin resistant BFCN by culturing them with high insulin concentrations. By means of these paradigms, we observed that nerve growth factor (NGF) reduces insulin resistance in vitro and in vivo. NGF activates the insulin receptor substrate 1 (IRS1) and rescues c-Fos expression and glucose metabolism. This effect involves binding of activated IRS1 to the NGF receptor TrkA, and is lost in presence of the specific IRS inhibitor NT157. Overall, our findings indicate that, in a well-established animal model of AD, the medial septum develops insulin resistance several months before it is detectable in the neocortex and hippocampus. Remarkably, NGF counteracts molecular alterations downstream of insulin-resistant receptor and its nasal administration restores insulin signaling in 3×Tg-AD mice by TrkA/IRS1 activation. The cross-talk between NGF and insulin pathways downstream the insulin receptor suggests novel potential therapeutic targets to slow cognitive decline in AD and diabetes-related brain insulin resistance.


Alzheimer Disease/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , Insulin/pharmacology , Nerve Growth Factor/pharmacology , Septal Nuclei/metabolism , Alzheimer Disease/genetics , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , Phosphorylation/drug effects , Rats , Rats, Wistar , Receptor, Insulin/metabolism , Septal Nuclei/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(2): 509-519, 2018 Feb.
Article En | MEDLINE | ID: mdl-29154925

Amyotrophic Lateral Sclerosis (ALS) is a chronic neurodegenerative disease affecting upper and lower motor neurons, with unknown aetiology. Lipid rafts, cholesterol enriched microdomains of the plasma membrane, have been linked to neurodegenerative disorders like ALS. The NMDA-receptor subcellular localization in lipid rafts is known to play many roles, from modulating memory strength to neurotoxicity. In this study, performed on the widely used G93A mouse model of ALS, we have shown an equal content of total membrane cholesterol in Control and G93A cortical cultures. Moreover, by electrophysiological studies, we have recorded NMDA- and AMPA-evoked currents which were not significantly different between the two neuronal populations. To study the role of membrane cholesterol on glutamate receptor functionality, we have analysed NMDA and AMPA receptors following cholesterol membrane depletion by methyl-ß-cyclodextrin (MßCD). Interestingly, MßCD chronic treatment has provoked a significant reduction of NMDA-evoked currents in both cellular populations which was dose- and time-dependent but significantly higher in ALS neurons compared to Control. The different MßCD effect on NMDA-evoked currents was not due to a different membrane receptor subunit composition but seemed to cause in both neuronal populations a NMDA receptor membrane redistribution. MßCD treatment effect was receptor-specific since no alterations in the two neuronal populations were detected on AMPA receptors. These results lead us to speculate for an altered proteomic composition of lipid rafts in cortical mutated neurons and suggest the need for further studies on the lipid rafts composition and on their interaction with membrane receptors in ALS cortices.


Amyotrophic Lateral Sclerosis/metabolism , Cholesterol/metabolism , Membrane Microdomains/chemistry , Motor Neurons/cytology , N-Methylaspartate/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cell Membrane/metabolism , Cell Survival , Disease Models, Animal , Electrophysiology , Female , Glutamic Acid/metabolism , Humans , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Proteomics , Receptors, AMPA/metabolism , Signal Transduction , Superoxide Dismutase-1/genetics , beta-Cyclodextrins/pharmacology
10.
Int J Mol Sci ; 18(6)2017 Jun 20.
Article En | MEDLINE | ID: mdl-28632177

Dysfunction of nerve growth factor (NGF) and its high-affinity Tropomyosin receptor kinase A (TrkA) receptor has been suggested to contribute to the selective degeneration of basal forebrain cholinergic neurons (BFCN) associated with the progressive cognitive decline in Alzheimer's disease (AD). The aim of this review is to describe our progress in elucidating the molecular mechanisms underlying the dynamic interplay between NGF/TrkA signaling and amyloid precursor protein (APP) metabolism within the context of AD neuropathology. This is mainly based on the finding that TrkA receptor binding to APP depends on a minimal stretch of ~20 amino acids located in the juxtamembrane/extracellular domain of APP that carries the α- and ß-secretase cleavage sites. Here, we provide evidence that: (i) NGF could be one of the "routing" proteins responsible for modulating the metabolism of APP from amyloidogenic towards non-amyloidogenic processing via binding to the TrkA receptor; (ii) the loss of NGF/TrkA signaling could be linked to sporadic AD contributing to the classical hallmarks of the neuropathology, such as synaptic loss, ß-amyloid peptide (Aß) deposition and tau abnormalities. These findings will hopefully help to design therapeutic strategies for AD treatment aimed at preserving cholinergic function and anti-amyloidogenic activity of the physiological NGF/TrkA pathway in the septo-hippocampal system.


Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Signal Transduction , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins , Animals , Cholinergic Neurons , Hippocampus/metabolism , Humans , Neuropathology , Synapses/metabolism , tau Proteins/metabolism
11.
PLoS One ; 12(3): e0173484, 2017.
Article En | MEDLINE | ID: mdl-28267767

The early phase of life represents a critical period for the development of an organism. Interestingly, early life experiences are able to influence the development of the gastrointestinal tract and the reactivity to colonic inflammatory stress. We recently demonstrated that adult male rats exposed to low doses of corticosterone during lactation (CORT-nursed rats) are protected against experimental colitis induced by the intracolonic infusion of 2,4,6-trinitrobenzenesulfonic acid (TNBS). Based on these interesting results, we wanted to better investigate which cellular actors could be involved in the protection of CORT-nursed rats from TNBS-induced experimental colitis. Therefore, in the present work, we focused our attention on different factors implicated in GR-mediated anti-inflammatory effect. To address this issue, colonic tissues, collected from control and CORT-nursed healthy animals and from control and CORT-nursed colitic rats, were processed and the following inflammatory factors were evaluated: the expression of (i) glucocorticoid receptors (GR), (ii) glucocorticoid-induced leucine zipper (GILZ), (iii) phospho-p65NF-κB, (iv) the pro-inflammatory cytokines IL-1ß and TNF-α, (v) the prokineticins PK2 and PK2L and (vi) their receptors PKR1 and PKR2. We found that adult CORT-nursed rats, in comparison to controls, showed increased expression of colonic GR and reduced expression of pro-inflammatory molecules (IL-1ß, TNF-α, PK2 and PK2L) in response to inflammatory colitis. The observed changes were associated with an increase in GILZ colonic expression and with a reduction in phospo-p65NF-κB colonic expression.


Colitis/etiology , Colitis/metabolism , Corticosterone/administration & dosage , Gastrointestinal Hormones/metabolism , Lactation/drug effects , Maternal Exposure , Receptors, Glucocorticoid/metabolism , Animals , Breast Feeding , Colitis/pathology , Colitis/prevention & control , Disease Models, Animal , Female , Gene Expression , Male , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Glucocorticoid/genetics , Trinitrobenzenesulfonic Acid/adverse effects
12.
Front Mol Neurosci ; 10: 15, 2017.
Article En | MEDLINE | ID: mdl-28197073

The amyloid precursor protein (APP) interacts with the tropomyosin receptor kinase A (TrkA) in normal rat, mouse, and human brain tissue but not in Alzheimer's disease (AD) brain tissue. However, it has not been reported whether the two proteins interact directly, and if so, which domains are involved. Clarifying these points will increase our understanding of the role and regulation of the TrkA/APP interaction in normal brain functioning as well as in AD. Here we addressed these questions using bimolecular fluorescence complementation (BiFC) and the proximity ligation assay (PLA). We demonstrated that exogenously expressed APP and TrkA associate through their juxtamembrane/transmembrane domains, to form a complex that localizes mainly to the plasma membrane, endoplasmic reticulum (ER) and Golgi. Formation of the complex was inhibited by p75NTR, ShcC and Mint-2. Importantly, we demonstrated that the association between endogenous APP and TrkA in primary septal neurons were modified by NGF, or by drugs that either inhibit ER-to-Golgi transport or perturb microtubules and microfilaments. Interestingly, several agents that induce cell death [amyloid ß (Aß)-peptide, staurosporine and rapamycin], albeit via different mechanisms, all caused dissociation of APP/TrkA complexes and increased production of C-terminal fragment (ß-CTF) APP fragment. These findings open new perspectives for investigating the interplay between these proteins during neurodegeneration and AD.

13.
Aging Cell ; 15(4): 661-72, 2016 08.
Article En | MEDLINE | ID: mdl-27076121

NGF has been implicated in forebrain neuroprotection from amyloidogenesis and Alzheimer's disease (AD). However, the underlying molecular mechanisms are still poorly understood. Here, we investigated the role of NGF signalling in the metabolism of amyloid precursor protein (APP) in forebrain neurons using primary cultures of septal neurons and acute septo-hippocampal brain slices. In this study, we show that NGF controls the basal level of APP phosphorylation at Thr668 (T668) by downregulating the activity of the Ser/Thr kinase JNK(p54) through the Tyr kinase signalling adaptor SH2-containing sequence C (ShcC). We also found that the specific NGF receptor, Tyr kinase A (TrkA), which is known to bind to APP, fails to interact with the fraction of APP molecules phosphorylated at T668 (APP(pT668) ). Accordingly, the amount of TrkA bound to APP is significantly reduced in the hippocampus of ShcC KO mice and of patients with AD in which elevated APP(pT668) levels are detected. NGF promotes TrkA binding to APP and APP trafficking to the Golgi, where APP-BACE interaction is hindered, finally resulting in reduced generation of sAPPß, CTFß and amyloid-beta (1-42). These results demonstrate that NGF signalling directly controls basal APP phosphorylation, subcellular localization and BACE cleavage, and pave the way for novel approaches specifically targeting ShcC signalling and/or the APP-TrkA interaction in AD therapy.


Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Down-Regulation/drug effects , Nerve Growth Factor/pharmacology , Phosphothreonine/metabolism , Adult , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Enzyme Activation/drug effects , Gene Deletion , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Hippocampus/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Phosphorylation/drug effects , Protein Binding/drug effects , Rats , Receptor, trkA/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism
14.
J Cell Sci ; 129(4): 804-16, 2016 Feb 15.
Article En | MEDLINE | ID: mdl-26743087

Disconnection between membrane signalling and actin networks can have catastrophic effects depending on cell size and polarity. The survival motor neuron (SMN) protein is ubiquitously involved in assembly of spliceosomal small nuclear ribonucleoprotein particles. Other SMN functions could, however, affect cellular activities driving asymmetrical cell surface expansions. Genes able to mitigate SMN deficiency operate within pathways in which SMN can act, such as mRNA translation, actin network and endocytosis. Here, we found that SMN accumulates at membrane protrusions during the dynamic rearrangement of the actin filaments. In addition to localization data, we show that SMN interacts with caveolin-1, which mediates anchoring of translation machinery components. Importantly, SMN deficiency depletes the plasma membrane of ribosomes, and this correlates with the failure of fibroblasts to extend membrane protrusions. These findings strongly support a relationship between SMN and membrane dynamics. We propose that SMN could assembly translational platforms associated with and governed by the plasma membrane. This activity could be crucial in cells that have an exacerbated interdependence of membrane remodelling and local protein synthesis.


Cell Membrane/metabolism , SMN Complex Proteins/physiology , Actin Cytoskeleton/metabolism , Caveolin 1/metabolism , Cell Membrane/ultrastructure , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Cells, Cultured , Humans , Protein Biosynthesis , Protein Transport , Ribosomes/metabolism
15.
Methods Mol Biol ; 1254: 115-28, 2015.
Article En | MEDLINE | ID: mdl-25431061

Double-stranded RNA -mediated interference (RNAi ) is a new simple and fast research tool for shutting down genes and characterizes function of their respective proteins. Many strategies for design and delivery of siRNA to target cells are available. Here, we describe the use of lentiviral short hairpin RNA (shRNA) RNA silencing to identify the involvement of d-serine racemase (SR )- an enzyme that syntheses d-serine to modulate glutamate- N-methyl-D-aspartate receptor- in regulating rat cerebellar granule neurons (CGN ) apoptosis. Apoptosis is induced by serum and KCl withdrawal and is detected with fluorometric caspase 3 assay.


Apoptosis/genetics , Neurons/metabolism , RNA, Small Interfering/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Humans , Lentivirus/genetics , Molecular Biology/methods , Rats
16.
Article En | MEDLINE | ID: mdl-24795622

A fine balance between cell survival and cell death is required to sculpt the nervous system during development. However, an excess of cell death can occur following trauma, exposure to neurotoxins or alcohol, and some developmental and neurodegenerative diseases, such as Alzheimer's disease (AD). N-Methyl-D-aspartate receptors (NMDARs) support synaptic plasticity and survival of many neuronal populations whereas inappropriate activation may promote various forms of cell death, apoptosis, and necrosis representing the two extremes of a continuum of cell death processes both "in vitro" and "in vivo." Hence, by identifying the switches controlling pro-survival vs. apoptosis and apoptosis vs. pro-excitotoxic outcome of NMDAR stimulation, NMDAR modulators could be developed that selectively block the cell death enhancing pro-survival signaling or synaptic plasticity mediated by NMDAR. Among these modulators, a role is emerging for the enzyme serine racemase (SR) that synthesizes D-serine, a key co-agonist with glutamate at NMDAR. This review summarizes the experimental evidence from "in vitro" neuronal cultures-with special emphasis on cerebellar granule neurons (CGNs)-and "in vivo" models of neurodegeneration, where the dual role of the SR/D-serine pathway as a master regulator of apoptosis and the apoptosis-necrosis shift will be discussed.

17.
Neurobiol Aging ; 34(11): 2551-63, 2013 Nov.
Article En | MEDLINE | ID: mdl-23769395

Different pathological tau species are involved in memory loss in Alzheimer's disease, the most common cause of dementia among older people. However, little is known about how tau pathology directly affects adult hippocampal neurogenesis, a unique form of structural plasticity implicated in hippocampus-dependent spatial learning and mood-related behavior. To this aim, we generated a transgenic mouse model conditionally expressing a pathological tau fragment (26-230 aa of the longest human tau isoform, or N-tau) in nestin-positive stem/progenitor cells. We found that N-tau reduced the proliferation of progenitor cells in the adult dentate gyrus, reduced cell survival and increased cell death by a caspase-3-independent mechanism, and recruited microglia. Although the number of terminally differentiated neurons was reduced, these showed an increased dendritic arborization and spine density. This resulted in an increase of anxiety-related behavior and an impairment of episodic-like memory, whereas less complex forms of spatial learning remained unaltered. Understanding how pathological tau species directly affect neurogenesis is important for developing potential therapeutic strategies to direct neurogenic instructive cues for hippocampal function repair.


Anxiety/genetics , Hippocampus/physiopathology , Memory Disorders/genetics , Neurogenesis/genetics , tau Proteins/metabolism , Adult Stem Cells/drug effects , Adult Stem Cells/physiology , Animals , Dark Adaptation/genetics , Dendrites/pathology , Dendrites/ultrastructure , Disease Models, Animal , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Genetic Vectors/physiology , Hippocampus/pathology , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nestin/genetics , Time Factors , beta-Galactosidase/metabolism , tau Proteins/genetics
18.
J Alzheimers Dis ; 33(3): 767-73, 2013.
Article En | MEDLINE | ID: mdl-23001708

Diabetes often correlates with tau phosphorylation and the development of Alzheimer's disease. Both are associated with brain cholinergic dysfunction that could benefit from nerve growth factor (NGF)-based therapies. Electroacupuncture (EA) improves brain NGF availability and action. Here we assessed the variations of NGF and tau phosphorylation in the cortex and hippocampus, as well as the expression of choline acetyltransferase in the basal forebrain following diabetes induction and EA in adult rats. We found that EA counteracts diabetes-associated tau hyperphosphorylation and decreases in NGF and choline acetyltransferase, suggesting a possible beneficial effect of EA on brain cholinergic system in diabetes.


Brain/metabolism , Choline O-Acetyltransferase/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Electroacupuncture , tau Proteins/metabolism , Analysis of Variance , Animals , Brain/anatomy & histology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Expression Regulation/radiation effects , Glycogen Synthase Kinase 3/metabolism , Nerve Growth Factor/metabolism , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Receptor, trkA/metabolism
19.
Exp Neurol ; 247: 349-58, 2013 Sep.
Article En | MEDLINE | ID: mdl-23142186

Voltage-gated Ca(2+) channels (VGCCs) mediate calcium entry into neuronal cells in response to membrane depolarisation and play an essential role in a variety of physiological processes. In Amyotrophic Lateral Sclerosis (ALS), a fatal neurodegenerative disease caused by motor neuron degeneration in the brain and spinal cord, intracellular calcium dysregulation has been shown, while no studies have been carried out on VGCCs. Here we show that the subtype N-type Ca(2+) channels are over expressed in G93A cultured cortical neurons and in motor cortex of G93A mice compared to Controls. In fact, by western blotting, immunocytochemical and electrophysiological experiments, we observe higher membrane expression of N-type Ca(2+) channels in G93A neurons compared to Controls. G93A cortical neurons filled with calcium-sensitive dye Fura-2, show a net calcium entry during membrane depolarization that is significantly higher compared to Control. Analysis of neuronal vitality following the exposure of neurons to a high K(+) concentration (25 mM, 5h), shows a significant reduction of G93A cellular survival compared to Controls. N-type channels are involved in the G93A higher mortality because ω-conotoxin GVIA (1 µM), which selectively blocks these channels, is able to abolish the higher G93A mortality when added to the external medium. These data provide robust evidence for an excess of N-type Ca(2+) expression in G93A cortical neurons which induces a higher mortality following membrane depolarization. These results may be central to the understanding of pathogenic pathways in ALS and provide novel molecular targets for the design of rational therapies for the ALS disorder.


Amyotrophic Lateral Sclerosis/pathology , Calcium Channels, N-Type/metabolism , Cerebral Cortex/pathology , Motor Neurons/metabolism , Animals , Animals, Newborn , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/genetics , Cell Survival , Cells, Cultured , Cerebral Cortex/cytology , Cytophotometry , Disease Models, Animal , Electric Stimulation , Gene Expression Regulation/drug effects , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Motor Neurons/pathology , Patch-Clamp Techniques , Sodium Channel Blockers/pharmacology , Superoxide Dismutase/immunology , Superoxide Dismutase/toxicity , Tetrodotoxin/pharmacology , omega-Agatoxin IVA/pharmacology , omega-Conotoxin GVIA/pharmacology
20.
Aging Cell ; 11(6): 1084-93, 2012 Dec.
Article En | MEDLINE | ID: mdl-23020178

Processing of Aß-precursor protein (APP) plays an important role in Alzheimer's disease (AD) pathogenesis. The APP intracellular domain contains residues important in regulating APP function and processing, in particular the 682YENPTY687 motif. To dissect the functions of this sequence in vivo, we created an APP knock-in allele mutating Y682 to Gly (APP(YG/YG) mice). This mutation alters the processing of APP and TrkA signaling and leads to postnatal lethality and neuromuscular synapse defects when expressed on an APP-like protein 2 KO background. This evidence prompted us to characterize further the APP(YG/YG) mice. Here, we show that APP(YG/YG) mice develop aging-dependent decline in cognitive and neuromuscular functions, a progressive reduction in dendritic spines, cholinergic tone, and TrkA levels in brain regions governing cognitive and motor functions. These data are consistent with our previous findings linking NGF and APP signaling and suggest a causal relationship between altered synaptic connectivity, cholinergic tone depression and TrkA signaling deficit, and cognitive and neuromuscular decline in APP(YG/YG) mice. The profound deficits caused by the Y682 mutation underscore the biological importance of APP and indicate that APP(YG/YG) are a valuable mouse model to study APP functions in physiological and pathological processes.


Aging/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Motor Endplate/metabolism , Synapses/metabolism , Tyrosine/metabolism , Aging/genetics , Aging/pathology , Amino Acid Motifs , Amino Acid Substitution , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal , Brain/pathology , Cognition , Gene Expression Regulation , Gene Knock-In Techniques , Glycine/genetics , Glycine/metabolism , Mice , Mice, Transgenic , Motor Activity , Motor Endplate/pathology , Protein Structure, Tertiary , Receptor, trkA/genetics , Receptor, trkA/metabolism , Signal Transduction , Synapses/pathology , Tyrosine/genetics
...