Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Curr Neuropharmacol ; 2023 08 17.
Article in English | MEDLINE | ID: mdl-37592816

ABSTRACT

Autism spectrum disorder (ASD) includes a heterogeneous group of complex neurodevel opmental disorders characterized by atypical behaviors with two core pathological manifestations: deficits in social interaction/communication and repetitive behaviors, which are associated with disturbed redox homeostasis. Modulation of cellular resilience mechanisms induced by low levels of stressors represents a novel approach for the development of therapeutic strategies, and in this context, neuroprotective effects of a wide range of polyphenol compounds have been demonstrated in several in vitro and in vivo studies and thoroughly reviewed by [2, 3]. Mushrooms have been used in traditional medicine for many years and have been associated with a long list of therapeutic properties, including antitumor, immunomodulatory, antioxidant, antiviral, antibacterial, and hepatoprotective effects [4]. Our recent studies have strikingly indicated the presence of polyphenols in nutritional mushrooms and demonstrated their protective effects in different models of neurodegenerative disorders in humans and rats [5, 6]. Although their therapeutic effects are exerted through multiple mechanisms, increasing attention is focusing on their capacity to induce endogenous defense systems by modulating cellular signaling processes, such as nuclear factor erythroid 2 related factor 2 (Nrf2) and nuclear factor-kappa B (NF-κB) pathways. Here we discuss the protective role of hormesis and its modulation by hormetic nutrients in ASD.

2.
Free Radic Biol Med ; 193(Pt 2): 657-668, 2022 11 20.
Article in English | MEDLINE | ID: mdl-36400326

ABSTRACT

The nitric oxide (NO)/cGMP pathway has been extensively studied for its pivotal role in synaptic plasticity and memory processes, resulting in an increase of cAMP response element-binding (CREB) phosphorylation, and consequent synthesis of plasticity-related proteins. The NO/cGMP/CREB signaling is downregulated during aging and neurodegenerative disorders and is affected by Amyloid-ß peptide (Aß) and tau protein, whose increase and deposition is considered the key pathogenic event of Alzheimer's disease (AD). On the other hand, in physiological conditions, the crosstalk between the NO/cGMP/PKG/CREB pathway and Aß ensures long-term potentiation and memory formation. This review summarizes the current knowledge on the interaction between the NO/cGMP/PKG/CREB pathway and Aß in the healthy and diseased brain, offering a new perspective to shed light on AD pathophysiology. We will focus on the synaptic mechanisms underlying Aß physiological interplay with cGMP pathway and how this balance is corrupted in AD, as high levels of Aß interfere with NO production and cGMP molecular signaling leading to cognitive impairment. Finally, we will discuss results from preclinical and clinical studies proposing the increase of cGMP signaling as a therapeutic strategy in the treatment of AD.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/genetics , Nitric Oxide , Amyloid beta-Peptides/genetics , Signal Transduction , Cyclic GMP
3.
Front Pharmacol ; 10: 693, 2019.
Article in English | MEDLINE | ID: mdl-31293421

ABSTRACT

Depression is a risk factor for the development of Alzheimer's disease (AD), and the presence of depressive symptoms significantly increases the conversion of mild cognitive impairment (MCI) into AD. A long-term treatment with antidepressants reduces the risk to develop AD, and different second-generation antidepressants such as selective serotonin reuptake inhibitors (SSRIs) are currently being studied for their neuroprotective properties in AD. In the present work, the SSRI fluoxetine and the new multimodal antidepressant vortioxetine were tested for their ability to prevent memory deficits and depressive-like phenotype induced by intracerebroventricular injection of amyloid-ß (1-42) (Aß1-42) oligomers in 2-month-old C57BL/6 mice. Starting from 7 days before Aß injection, fluoxetine (10 mg/kg) and vortioxetine (5 and 10 mg/kg) were intraperitoneally injected daily for 24 days. Chronic treatment with fluoxetine and vortioxetine (both at the dose of 10 mg/kg) was able to rescue the loss of memory assessed 14 days after Aß injection by the passive avoidance task and the object recognition test. Both antidepressants reversed the increase in immobility time detected 19 days after Aß injection by forced swim test. Vortioxetine exerted significant antidepressant effects also at the dose of 5 mg/kg. A significant deficit of transforming growth factor-ß1 (TGF-ß1), paralleling memory deficits and depressive-like phenotype, was found in the hippocampus of Aß-injected mice in combination with a significant reduction of the synaptic proteins synaptophysin and PSD-95. Fluoxetine and vortioxetine completely rescued hippocampal TGF-ß1 levels in Aß-injected mice as well as synaptophysin and PSD-95 levels. This is the first evidence that a chronic treatment with fluoxetine or vortioxetine can prevent both cognitive deficits and depressive-like phenotype in a non-transgenic animal model of AD with a key contribution of TGF-ß1.

4.
Cell Signal ; 62: 109338, 2019 10.
Article in English | MEDLINE | ID: mdl-31176021

ABSTRACT

α7 nicotinic acetylcholine receptors (α7nAChRs) have been targeted to improve cognition in different neurological and psychiatric disorders. Nevertheless, no α7nAChR activating ligand has been clinically approved. Here, we investigated the effects of antagonizing α7nAChRs using the selective antagonist methyllycaconitine (MLA) on receptor activity in vitro and cognitive functioning in vivo. Picomolar concentrations of MLA significantly potentiated receptor responses in electrophysiological experiments mimicking the in vivo situation. Furthermore, microdialysis studies showed that MLA administration substantially increased hippocampal glutamate efflux which is related to memory processes. Accordingly, pre-tetanus administration of low MLA concentrations produced longer lasting potentiation (long-term potentiation, LTP) in studies examining hippocampal plasticity. Moreover, low doses of MLA improved acquisition, but not consolidation memory processes in rats. While the focus to enhance cognition by modulating α7nAChRs lies on agonists and positive modulators, antagonists at low doses should provide a novel approach to improve cognition in neurological and psychiatric disorders.


Subject(s)
Aconitine/analogs & derivatives , Cognition/drug effects , Memory/drug effects , alpha7 Nicotinic Acetylcholine Receptor/genetics , Aconitine/metabolism , Aconitine/pharmacology , Animals , Cognition/physiology , Disease Models, Animal , Glutamic Acid/genetics , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Memory/physiology , Nicotinic Antagonists/pharmacology , Rats , Receptors, Nicotinic/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
5.
J Neurosci ; 39(30): 5986-6000, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31127002

ABSTRACT

Failure of anti-amyloid-ß peptide (Aß) therapies against Alzheimer's disease (AD), a neurodegenerative disorder characterized by high amounts of the peptide in the brain, raised the question of the physiological role of Aß released at low concentrations in the healthy brain. To address this question, we studied the presynaptic and postsynaptic mechanisms underlying the neuromodulatory action of picomolar amounts of oligomeric Aß42 (oAß42) on synaptic glutamatergic function in male and female mice. We found that 200 pm oAß42 induces an increase of frequency of miniature EPSCs and a decrease of paired pulse facilitation, associated with an increase in docked vesicle number, indicating that it augments neurotransmitter release at presynaptic level. oAß42 also produced postsynaptic changes as shown by an increased length of postsynaptic density, accompanied by an increased expression of plasticity-related proteins such as cAMP-responsive element binding protein phosphorylated at Ser133, calcium-calmodulin-dependent kinase II phosphorylated at Thr286, and brain-derived neurotrophic factor, suggesting a role for Aß in synaptic tagging. These changes resulted in the conversion of early into late long-term potentiation through the nitric oxide/cGMP/protein kinase G intracellular cascade consistent with a cGMP-dependent switch from short- to long-term memory observed in vivo after intrahippocampal administration of picomolar amounts of oAß42 These effects were present upon extracellular but not intracellular application of the peptide and involved α7 nicotinic acetylcholine receptors. These observations clarified the physiological role of oAß42 in synaptic function and memory formation providing solid fundamentals for investigating the pathological effects of high Aß levels in the AD brains.SIGNIFICANCE STATEMENT High levels of oligomeric amyloid-ß42 (oAß42) induce synaptic dysfunction leading to memory impairment in Alzheimer's disease (AD). However, at picomolar concentrations, the peptide is needed to ensure long-term potentiation (LTP) and memory. Here, we show that extracellular 200 pm oAß42 concentrations increase neurotransmitter release, number of docked vesicles, postsynaptic density length, and expression of plasticity-related proteins leading to the conversion of early LTP into late LTP and of short-term memory into long-term memory. These effects require α7 nicotinic acetylcholine receptors and are mediated through the nitric oxide/cGMP/protein kinase G pathway. The knowledge of Aß function in the healthy brain might be useful to understand the causes leading to its increase and detrimental effect in AD.


Subject(s)
Amyloid beta-Peptides/administration & dosage , Extracellular Fluid/physiology , Memory/physiology , Neurotransmitter Agents/administration & dosage , Peptide Fragments/administration & dosage , Presynaptic Terminals/physiology , Synapses/physiology , Animals , Extracellular Fluid/drug effects , Female , Hippocampus/drug effects , Hippocampus/physiology , Injections, Intraventricular , Male , Memory/drug effects , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Presynaptic Terminals/drug effects , Rats , Rats, Wistar , Synapses/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
7.
Neurobiol Aging ; 71: 51-60, 2018 11.
Article in English | MEDLINE | ID: mdl-30092511

ABSTRACT

The increase of oligomeric amyloid-beta (oAß) has been related to synaptic dysfunction, thought to be the earliest event in Alzheimer's disease pathophysiology. Conversely, the suppression of endogenous Aß impaired synaptic plasticity and memory, suggesting that the peptide is needed in the healthy brain. However, different species, aggregation forms and concentrations of Aß might differently influence synaptic function/dysfunction. Here, we have tested the contribution of monomeric and oligomeric Aß42 and Aß40 at 200 nM and 200 pM concentrations on hippocampal long-term potentiation and spatial memory. We found that, when at 200 nM, oAß40, oAß42, and monomeric Aß42 impaired long-term potentiation and memory, whereas only oAß42 200 pM enhanced synaptic plasticity and memory and rescued the detrimental effect due to depletion of endogenous Aß. Interestingly, quantification of monomer-like and oligomer-like species carried out by transmission electron microscopy revealed an increase of the monomer/oligomer ratio in the oAß42 200 pM preparation, suggesting that the content of monomers and oligomers depends on the final concentration of the solution.


Subject(s)
Amyloid beta-Peptides/physiology , Hippocampus/physiology , Long-Term Potentiation , Peptide Fragments/physiology , Spatial Memory/physiology , Amyloid beta-Peptides/administration & dosage , Animals , Female , Hippocampus/drug effects , Humans , Long-Term Potentiation/drug effects , Male , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Protein Aggregates , Protein Isoforms/administration & dosage , Protein Isoforms/physiology , Spatial Memory/drug effects
8.
Neuropharmacology ; 138: 151-159, 2018 08.
Article in English | MEDLINE | ID: mdl-29885420

ABSTRACT

Cyclic nucleotides cAMP and cGMP cooperate to ensure memory acquisition and consolidation. Increasing their levels by phosphodiesterase inhibitors (PDE-Is) enhanced cognitive functions and rescued memory loss in different models of aging and Alzheimer's disease (AD). However, side effects due to the high doses used limited their application in humans. Based on previous studies suggesting that combinations of sub-efficacious doses of cAMP- and cGMP-specific PDE-Is improved synaptic plasticity and memory in physiological conditions, here we aimed to study whether this treatment was effective to counteract the AD phenotype in APPswe mice. We found that a 3-week chronic treatment with a combination of sub-efficacious doses of the cAMP-specific PDE4-I roflumilast (0.01 mg/kg) and the cGMP-specific PDE5-I vardenafil (0.1 mg/kg) improved recognition, spatial and contextual fear memory. Importantly, the cognitive enhancement persisted for 2 months beyond administration. This long-lasting action, and the possibility to minimize side effects due to the low doses used, might open feasible therapeutic strategies against AD.


Subject(s)
Alzheimer Disease/drug therapy , Aminopyridines/pharmacology , Benzamides/pharmacology , Memory/drug effects , Nootropic Agents/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Vardenafil Dihydrochloride/pharmacology , Alzheimer Disease/enzymology , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Cyclopropanes/pharmacology , Disease Models, Animal , Female , Humans , Male , Memory/physiology , Mice, Transgenic , Random Allocation
9.
J Alzheimers Dis ; 64(s1): S611-S631, 2018.
Article in English | MEDLINE | ID: mdl-29865055

ABSTRACT

The "Amyloid Cascade Hypothesis" has dominated the Alzheimer's disease (AD) field in the last 25 years. It posits that the increase of amyloid-ß (Aß) is the key event in AD that triggers tau pathology followed by neuronal death and eventually, the disease. However, therapeutic approaches aimed at decreasing Aß levels have so far failed, and tau-based clinical trials have not yet produced positive findings. This begs the question of whether the hypothesis is correct. Here we have examined literature on the role of Aß and tau in synaptic dysfunction, memory loss, and seeding and spreading of AD, highlighting important parallelisms between the two proteins in all of these phenomena. We discuss novel findings showing binding of both Aß and tau oligomers to amyloid-ß protein precursor (AßPP), and the requirement for the presence of this protein for both Aß and tau to enter neurons and induce abnormal synaptic function and memory. Most importantly, we propose a novel view of AD pathogenesis in which extracellular oligomers of Aß and tau act in parallel and upstream of AßPP. Such a view will call for a reconsideration of therapeutic approaches directed against Aß and tau, paving the way to an increased interest toward AßPP, both for understanding the pathogenesis of the disease and elaborating new therapeutic strategies.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Alzheimer Disease/therapy , Animals , Humans , Signal Transduction
10.
Elife ; 62017 07 11.
Article in English | MEDLINE | ID: mdl-28696204

ABSTRACT

The concurrent application of subtoxic doses of soluble oligomeric forms of human amyloid-beta (oAß) and Tau (oTau) proteins impairs memory and its electrophysiological surrogate long-term potentiation (LTP), effects that may be mediated by intra-neuronal oligomers uptake. Intrigued by these findings, we investigated whether oAß and oTau share a common mechanism when they impair memory and LTP in mice. We found that as already shown for oAß, also oTau can bind to amyloid precursor protein (APP). Moreover, efficient intra-neuronal uptake of oAß and oTau requires expression of APP. Finally, the toxic effect of both extracellular oAß and oTau on memory and LTP is dependent upon APP since APP-KO mice were resistant to oAß- and oTau-induced defects in spatial/associative memory and LTP. Thus, APP might serve as a common therapeutic target against Alzheimer's Disease (AD) and a host of other neurodegenerative diseases characterized by abnormal levels of Aß and/or Tau.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Long-Term Potentiation , Memory Disorders/physiopathology , Neurons/physiology , Peptide Fragments/metabolism , Protein Multimerization , tau Proteins/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/toxicity , Protein Binding , tau Proteins/toxicity
11.
J Neurosci ; 37(29): 6926-6937, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28626017

ABSTRACT

High levels of amyloid-ß peptide (Aß) have been related to Alzheimer's disease pathogenesis. However, in the healthy brain, low physiologically relevant concentrations of Aß are necessary for long-term potentiation (LTP) and memory. Because cGMP plays a key role in these processes, here we investigated whether the cyclic nucleotide cGMP influences Aß levels and function during LTP and memory. We demonstrate that the increase of cGMP levels by the phosphodiesterase-5 inhibitors sildenafil and vardenafil induces a parallel release of Aß due to a change in the approximation of amyloid precursor protein (APP) and the ß-site APP cleaving enzyme 1. Moreover, electrophysiological and behavioral studies performed on animals of both sexes showed that blocking Aß function, by using anti-murine Aß antibodies or APP knock-out mice, prevents the cGMP-dependent enhancement of LTP and memory. Our data suggest that cGMP positively regulates Aß levels in the healthy brain which, in turn, boosts synaptic plasticity and memory.SIGNIFICANCE STATEMENT Amyloid-ß (Aß) is a key pathogenetic factor in Alzheimer's disease. However, low concentrations of endogenous Aß, mimicking levels of the peptide in the healthy brain, enhance hippocampal long-term potentiation (LTP) and memory. Because the second messenger cGMP exerts a central role in LTP mechanisms, here we studied whether cGMP affects Aß levels and function during LTP. We show that cGMP enhances Aß production by increasing the APP/BACE-1 convergence in endolysosomal compartments. Moreover, the cGMP-induced enhancement of LTP and memory was disrupted by blockade of Aß, suggesting that the physiological effect of the cyclic nucleotide on LTP and memory is dependent upon Aß.


Subject(s)
Amyloid beta-Peptides/metabolism , Cyclic GMP/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Memory/physiology , Mental Recall/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats, Sprague-Dawley , Task Performance and Analysis
12.
Eur J Pharmacol ; 817: 59-70, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28577963

ABSTRACT

The response of a biological system to an endogenous or exogenous molecule depends upon the dose. For this reason, performing dose-response curves is crucial to understand physiological and pathophysiological phenomena, and to predict the effect of a drug. Most of the studies in pharmacological research have been performed according to the classical threshold model, focusing on higher doses able to ensure a biological effect. However, recent evidences pointed out the need to investigate the effect of low doses. Indeed, several molecules behave in a hormetic fashion, i.e. low-doses stimulate whereas high-doses inhibit a biological response. This is particularly interesting in CNS, where several physiological molecules involved in neuronal transmission during learning and memory have shown a biphasic effect that might represent the link between physiology and pathology. In this review we will focus on cholinergic, glutamatergic and nitrinergic transmission, because of their central role in learning and memory and their impairment in neurodegenerative disorders such as Alzheimer's disease. Pre-clinical studies performed on healthy adult animals and aged animals, as well as transgenic animal models of AD, have suggested a biphasic DR for acetylcholine, glutamate and nitric oxide. This stresses the relevance to perform DR curves when studying the mechanisms underlying synaptic plasticity and memory, the pharmacological profile of cognitive-enhancing drugs acting on these systems, and the possibility to combine low/ineffective doses of drugs that might have additive/synergistic effects, reducing the unwanted side effects associated to the high doses.


Subject(s)
Drug Discovery , Memory/physiology , Neuronal Plasticity , Acetylcholine/physiology , Animals , Cognitive Dysfunction/drug therapy , Dose-Response Relationship, Drug , Drug Therapy, Combination , Glutamic Acid/physiology , Humans , Nitric Oxide/physiology
13.
Mol Cell Neurosci ; 81: 64-71, 2017 06.
Article in English | MEDLINE | ID: mdl-28038945

ABSTRACT

Cell adhesion molecules (CAMs) have a pivotal role in building and maintaining synaptic structures during brain development participating in axonal elongation and pathfinding, glial guidance of neuronal migration, as well as myelination. CAMs expression persists in the adult brain particularly in structures undergoing postnatal neurogenesis and involved in synaptic plasticity and memory as the hippocampus. Among the neural CAMs, we have recently focused on F3/Contactin, a glycosylphosphatidyl inositol-anchored glycoprotein belonging to the immunoglobulin superfamily, involved in neuronal development, synaptic maintenance and organization of neuronal networks. Here, we discuss our recent data suggesting that F3/Contactin exerts a role in hippocampal synaptic plasticity and memory in adult and aged mice. In particular, we have studied long-term potentiation (LTP), spatial and object recognition memory, and phosphorylation of the transcription factor cAMP-Responsive-Element Binding protein (CREB) in a transgenic mouse model of F3/Contactin overexpression. We also investigated whether F3/Contactin might influence neuronal apoptosis and the production of amyloid-beta peptide (Aß), known to be one of the main pathogenetic hallmarks of Alzheimer's disease (AD). In conclusion, a further understanding of F3/Contactin role in synaptic plasticity and memory might have interesting clinical outcomes in cognitive disorders, such as aging and AD, offering innovative therapeutic opportunities.


Subject(s)
Contactin 1/metabolism , Memory , Neuronal Plasticity , Animals , Brain/growth & development , Brain/metabolism , Brain/physiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Contactin 1/genetics , Humans
14.
Sci Rep ; 5: 37504, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27874066

ABSTRACT

Studies in vitro have demonstrated that ß3-adrenergic receptors (ß3-ARs) regulate protein metabolism in skeletal muscle by promoting protein synthesis and inhibiting protein degradation. In this study, we evaluated whether activation of ß3-ARs by the selective agonist CL316,243 modifies the functional and structural properties of skeletal muscles of healthy mice. Daily injections of CL316,243 for 15 days resulted in a significant improvement in muscle force production, assessed by grip strength and weight tests, and an increased myofiber cross-sectional area, indicative of muscle hypertrophy. In addition, atomic force microscopy revealed a significant effect of CL316,243 on the transversal stiffness of isolated muscle fibers. Interestingly, the expression level of mammalian target of rapamycin (mTOR) downstream targets and neuronal nitric oxide synthase (NOS) was also found to be enhanced in tibialis anterior and soleus muscles of CL316,243 treated mice, in accordance with previous data linking ß3-ARs to mTOR and NOS signaling pathways. In conclusion, our data suggest that CL316,243 systemic administration might be a novel therapeutic strategy worthy of further investigations in conditions of muscle wasting and weakness associated with aging and muscular diseases.


Subject(s)
Adrenergic beta-3 Receptor Agonists/pharmacology , Dioxoles/pharmacology , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Adrenergic beta-3 Receptor Agonists/administration & dosage , Animals , Dioxoles/administration & dosage , Gene Expression Regulation , Hypertrophy , Male , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Nitric Oxide Synthase Type I/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
15.
Sci Rep ; 6: 32553, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27581852

ABSTRACT

The oligomeric amyloid-ß (Aß) peptide is thought to contribute to the subtle amnesic changes in Alzheimer's disease (AD) by causing synaptic dysfunction. Here, we examined the time course of synaptic changes in mouse hippocampal neurons following exposure to Aß42 at picomolar concentrations, mimicking its physiological levels in the brain. We found opposite effects of the peptide with short exposures in the range of minutes enhancing synaptic plasticity, and longer exposures lasting several hours reducing it. The plasticity reduction was concomitant with an increase in the basal frequency of spontaneous neurotransmitter release, a higher basal number of functional presynaptic release sites, and a redistribution of synaptic proteins including the vesicle-associated proteins synapsin I, synaptophysin, and the post-synaptic glutamate receptor I. These synaptic alterations were mediated by cytoskeletal changes involving actin polymerization and p38 mitogen-activated protein kinase. These in vitro findings were confirmed in vivo with short hippocampal infusions of picomolar Aß enhancing contextual memory and prolonged infusions impairing it. Our findings provide a model for initiation of synaptic dysfunction whereby exposure to physiologic levels of Aß for a prolonged period of time causes microstructural changes at the synapse which result in increased transmitter release, failure of synaptic plasticity, and memory loss.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/pharmacology , Memory Disorders/diagnosis , Neuronal Plasticity/drug effects , Neurons/drug effects , Peptide Fragments/pharmacology , Synaptic Transmission/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Animals, Newborn , Disease Models, Animal , Gene Expression Regulation , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Primary Cell Culture , Protein Multimerization , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Synapses/drug effects , Synapsins/genetics , Synapsins/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
J Alzheimers Dis ; 52(1): 65-75, 2016 02 26.
Article in English | MEDLINE | ID: mdl-26967223

ABSTRACT

Rhodiola Rosea (R. Rosea) is a plant used in traditional popular medicine to enhance cognition and physical performance. R. Rosea medicinal properties have been related to its capability to act as an adaptogen, i.e., a substance able to increase the organism's resistance to a variety of chemical, biological, and physical stressors in a non-specific way. These adaptogen properties have been mainly attributed to the glycoside salidroside, one of the bioactive compounds present in the standardized extracts of R. Rosea. Here, we aimed to investigate whether a single dose of salidroside is able to affect memory and emotional behavior in wild type adult mice. We performed fear conditioning to assess cued and contextual memory, elevated plus maze and open field to evaluate anxiety, and tail suspension test to evaluate depression. Our results showed that a single i.p. administration of salidroside was able to enhance fear memory and exerted an anxiolytic and antidepressant effect. These data confirmed the adaptogenic effect of R. Rosea bioactive compounds in animal models and suggest that salidroside might represent an interesting pharmacological tool to ameliorate cognition and counteract mood disorders.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Glucosides/pharmacology , Nootropic Agents/pharmacology , Phenols/pharmacology , Animals , Anti-Anxiety Agents/chemistry , Antidepressive Agents/chemistry , Anxiety Disorders/drug therapy , Conditioning, Psychological/drug effects , Depressive Disorder/drug therapy , Disease Models, Animal , Drug Evaluation, Preclinical , Exploratory Behavior/drug effects , Fear/drug effects , Female , Freezing Reaction, Cataleptic/drug effects , Glucosides/chemistry , Male , Maze Learning/drug effects , Memory/drug effects , Mice, Inbred C57BL , Motor Activity/drug effects , Nootropic Agents/chemistry , Phenols/chemistry , Phytotherapy , Plant Extracts/chemistry , Rhodiola/chemistry
17.
Pharmacol Res ; 106: 10-20, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26875816

ABSTRACT

The ectopic re-activation of cell cycle in neurons is an early event in the pathogenesis of Alzheimer's disease (AD), which could lead to synaptic failure and ensuing cognitive deficits before frank neuronal death. Cytostatic drugs that act as cyclin-dependent kinase (CDK) inhibitors have been poorly investigated in animal models of AD. In the present study, we examined the effects of flavopiridol, an inhibitor of CDKs currently used as antineoplastic drug, against cell cycle reactivation and memory loss induced by intracerebroventricular injection of Aß1-42 oligomers in CD1 mice. Cycling neurons, scored as NeuN-positive cells expressing cyclin A, were found both in the frontal cortex and in the hippocampus of Aß-injected mice, paralleling memory deficits. Starting from three days after Aß injection, flavopiridol (0.5, 1 and 3mg/kg) was intraperitoneally injected daily, for eleven days. Here we show that a treatment with flavopiridol (0.5 and 1mg/kg) was able to rescue the loss of memory induced by Aß1-42, and to prevent the occurrence of ectopic cell-cycle events in the mouse frontal cortex and hippocampus. This is the first evidence that a cytostatic drug can prevent cognitive deficits in a non-transgenic animal model of AD.


Subject(s)
Amyloid beta-Peptides/adverse effects , Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Memory/drug effects , Peptide Fragments/adverse effects , Piperidines/pharmacology , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cognition Disorders/chemically induced , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Memory Disorders/etiology , Memory Disorders/metabolism , Mice , Neurons/drug effects , Neurons/metabolism
18.
Sci Rep ; 5: 11252, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26059637

ABSTRACT

Transforming Growth Factor ß1 (TGF-ß1) is a well-known neuroprotective and neurotrophic factor demonstrated to play a role in synaptic transmission. However, its involvement in physiological mechanisms underlying synaptic plasticity and memory at hippocampal level has not been thoroughly investigated. Here, we examine the role of TGF-ß1 in hippocampal long-term potentiation (LTP) and memory in adult wild type mice. Our data provide evidence that administration of exogenous TGF-ß1 is able to convert early-phase-LTP into late-phase-LTP. Furthermore, we show that the block of the endogenous TGF-ß1 signaling pathway by the specific TGF-ß1 inhibitor SB431542, impairs LTP and object recognition memory. The latter impairment was rescued by administration of exogenous TGF-ß1, suggesting that endogenously produced TGF-ß1 plays a role in physiological mechanisms underlying LTP and memory. Finally, TGF-ß1 functional effect correlates with an increased expression of the phosphorylated transcription factor cAMP-Responsive Element Binding protein.


Subject(s)
Hippocampus/metabolism , Memory , Neuronal Plasticity , Synapses/physiology , Transforming Growth Factor beta1/physiology , Animals , Mice , Mice, Inbred C57BL
19.
Expert Opin Drug Discov ; 10(7): 703-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25927677

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) is a neurodegenerative disorder characterized by memory loss and personality changes, leading to dementia. Histopathological hallmarks are represented by aggregates of beta-amyloid peptide (Aß) in senile plaques and deposition of hyperphosphorylated tau protein in neurofibrillary tangles in the brain. Rare forms of early onset familial Alzheimer's disease are due to gene mutations. This has prompted researchers to develop genetically modified animals that could recapitulate the main features of the disease. The use of these models is complemented by non-genetically modified animals. AREAS COVERED: This review summarizes the characteristics of the most used transgenic (Tg) and non-Tg models of AD. The authors have focused on models mainly used in their laboratories including amyloid precursor protein (APP) Tg2576, APP/presenilin 1, 3xAD, single h-Tau, non-Tg mice treated with acute injections of Aß or tau, and models of physiological aging. EXPERT OPINION: Animal models of disease might be very useful for studying the pathophysiology of the disease and for testing new therapeutics in preclinical studies but they do not reproduce the entire clinical features of human AD. When selecting a model, researchers should consider the various factors that might influence the phenotype. They should also consider the timing of testing/treating animals since the age at which each model develops certain aspects of the AD pathology varies.


Subject(s)
Alzheimer Disease/drug therapy , Disease Models, Animal , Drug Discovery/methods , Aging/physiology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Animals , Drug Evaluation, Preclinical/methods , Humans , Mice , Mice, Transgenic , Rodentia , Species Specificity
20.
Neurobiol Aging ; 36(4): 1702-1715, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25659859

ABSTRACT

We have recently shown that overexpression of the F3/contactin adhesive glycoprotein (also known as Contactin-1) promotes neurogenesis in adult hippocampus, which correlates with improved synaptic plasticity and memory. Because F3/contactin levels physiologically decrease with age, here, we aim at investigating whether its overexpression might counteract the cognitive decline in aged animals. For this we use 20- to 24-month-old TAG/F3 transgenic mice in which F3/contactin overexpression is driven by regulatory sequences from the gene encoding the transient axonal glycoprotein TAG-1 throughout development. We show that aged TAG/F3 mice display improved hippocampal long-term potentiation and memory compared with wild-type littermates. The same mice undergo a decrease of neuronal apoptosis at the hippocampal level, which correlated to a decrease of active caspase-3; by contrast, procaspase-3 and Bax as well as the anti-apoptotic and plasticity-related pathway BDNF/CREB/Bcl-2 were rather increased. Interestingly, amyloid-precursor protein processing was shifted toward sAPPα generation, with a decrease of sAPPß and amyloid-beta levels. Our data confirm that F3/contactin plays a role in hippocampal synaptic plasticity and memory also in aged mice, suggesting that it acts on molecular pathways related to apoptosis and amyloid-beta production.


Subject(s)
Aging/genetics , Contactin 1/genetics , Contactin 1/physiology , Hippocampus/physiology , Memory/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Aging/physiology , Aging/psychology , Amyloid beta-Peptides/metabolism , Animals , Apoptosis/genetics , Brain-Derived Neurotrophic Factor/physiology , Caspase 3/metabolism , Cognition Disorders/genetics , Gene Expression , Hippocampus/pathology , Long-Term Potentiation/genetics , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...