Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
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
2.
Pharmacol Res ; 141: 384-391, 2019 03.
Article in English | MEDLINE | ID: mdl-30648615

ABSTRACT

The dopamine D3 receptor (D3R), in the nucleus accumbens (NAc), plays an important role in alcohol reward mechanisms. The major neuronal type within the NAc is the GABAergic medium spiny neuron (MSN), whose activity is regulated by dopaminergic inputs. We previously reported that genetic deletion or pharmacological blockade of D3R increases GABAA α6 subunit in the ventral striatum. Here we tested the hypothesis that D3R-dependent changes in GABAA α6 subunit in the NAc affect voluntary alcohol intake, by influencing the inhibitory transmission of MSNs. We performed in vivo and ex vivo experiments in D3R knockout (D3R -/-) mice and wild type littermates (D3R +/+). Ro 15-4513, a high affinity α6-GABAA ligand was used to study α6 activity. At baseline, NAc α6 expression was negligible in D3R+/+, whereas it was robust in D3R-/-; other relevant GABAA subunits were not changed. In situ hybridization and qPCR confirmed α6 subunit mRNA expression especially in the NAc. In the drinking-in-the-dark paradigm, systemic administration of Ro 15-4513 inhibited alcohol intake in D3R+/+, but increased it in D3R-/-; this was confirmed by intra-NAc administration of Ro 15-4513 and furosemide, a selective α6-GABAA antagonist. Whole-cell patch-clamp showed peak amplitudes of miniature inhibitory postsynaptic currents in NAc medium spiny neurons higher in D3R-/- compared to D3R+/+; Ro 15-4513 reduced the peak amplitude in the NAc of D3R-/-, but not in D3R+/+. We conclude that D3R-dependent enhanced expression of α6 GABAA subunit inhibits voluntary alcohol intake by increasing GABA inhibition in the NAc.


Subject(s)
Binge Drinking/genetics , GABAergic Neurons/pathology , Receptors, Dopamine D3/genetics , Receptors, GABA-A/genetics , Animals , Binge Drinking/pathology , GABAergic Neurons/metabolism , Gene Expression Regulation , Male , Mice , Mice, Knockout , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Protein Subunits/genetics , RNA, Messenger/genetics
3.
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
4.
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
5.
Mol Cell Neurosci ; 81: 49-63, 2017 06.
Article in English | MEDLINE | ID: mdl-27871938

ABSTRACT

This review article focuses on the Contactin (CNTN) subset of the Immunoglobulin supergene family (IgC2/FNIII molecules), whose components share structural properties (the association of Immunoglobulin type C2 with Fibronectin type III domains), as well as a general role in cell contact formation and axonal growth control. IgC2/FNIII molecules include 6 highly related components (CNTN 1-6), associated with the cell membrane via a Glycosyl Phosphatidyl Inositol (GPI)-containing lipid tail. Contactin 1 and Contactin 2 share ~50 (49.38)% identity at the aminoacid level. They are components of the cell surface, from which they may be released in soluble forms. They bind heterophilically to multiple partners in cis and in trans, including members of the related L1CAM family and of the Neurexin family Contactin-associated proteins (CNTNAPs or Casprs). Such interactions are important for organising the neuronal membrane, as well as for modulating the growth and pathfinding of axon tracts. In addition, they also mediate the functional maturation of axons by promoting their interactions with myelinating cells at the nodal, paranodal and juxtaparanodal regions. Such interactions also mediate differential ionic channels (both Na+ and K+) distribution, which is of critical relevance in the generation of the peak-shaped action potential. Indeed, thanks to their interactions with Ankyrin G, Na+ channels map within the nodal regions, where they drive axonal depolarization. However, no ionic channels are found in the flanking Contactin1-containing paranodal regions, where CNTN1 interactions with Caspr1 and with the Ig superfamily component Neurofascin 155 in cis and in trans, respectively, build a molecular barrier between the node and the juxtaparanode. In this region K+ channels are clustered, depending upon molecular interactions with Contactin 2 and with Caspr2. In addition to these functions, the Contactins appear to have also a role in degenerative and inflammatory disorders: indeed Contactin 2 is involved in neurodegenerative disorders with a special reference to the Alzheimer disease, given its ability to work as a ligand of the Alzheimer Precursor Protein (APP), which results in increased Alzheimer Intracellular Domain (AICD) release in a γ-secretase-dependent manner. On the other hand Contactin 1 drives Notch signalling activation via the Hes pathway, which could be consistent with its ability to modulate neuroinflammation events, and with the possibility that Contactin 1-dependent interactions may participate to the pathogenesis of the Multiple Sclerosis and of other inflammatory disorders.


Subject(s)
Axons/metabolism , Contactins/metabolism , Neurodevelopmental Disorders/metabolism , Neurogenesis , Animals , Contactins/chemistry , Contactins/genetics , Humans , Neurodevelopmental Disorders/genetics
6.
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
7.
Brain ; 138(Pt 1): 203-16, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25472798

ABSTRACT

Alzheimer's disease is one of the most common causes of death worldwide, with poor treatment options. A tissue landmark of Alzheimer's disease is accumulation of the anomalous protein amyloid-ß in specific brain areas. Whether inflammation is an effect of amyloid-ß on the Alzheimer's disease brain, or rather it represents a cause for formation of amyloid plaques and intracellular tangles remains a subject of debate. TNFSF10, a proapoptotic cytokine of the TNF superfamily, is a mediator of amyloid-ß neurotoxicity. Here, we demonstrate that blocking TNFSF10 by administration of a neutralizing monoclonal antibody could attenuate the amyloid-ß-induced neurotoxicity in a triple transgenic mouse model of Alzheimer's disease (3xTg-AD). The effects of TNFSF10 neutralization on either cognitive parameters, as well as on the expression of TNFSF10, amyloid-ß, inflammatory mediators and GFAP were studied in the hippocampus of 3xTg-AD mice. Treatment with the TNFSF10 neutralizing antibody resulted in dramatic improvement of cognitive parameters, as assessed by the Morris water maze test and the novel object recognition test. These results were correlated with decreased protein expression of TNFSF10, amyloid-ß, inflammatory mediators and GFAP in the hippocampus. Finally, neutralization of TNFSF10 results in functional improvement and restrained immune/inflammatory response in the brain of 3xTg-AD mice in vivo. Thus, it is plausible to regard the TNFSF10 system as a potential target for efficacious treatment of amyloid-related disorders.


Subject(s)
Alzheimer Disease/drug therapy , Antibodies, Monoclonal/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/immunology , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cognition Disorders/etiology , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gliosis/drug therapy , Gliosis/etiology , Hippocampus/metabolism , Humans , Male , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics , Recognition, Psychology/drug effects , tau Proteins/genetics
8.
J Neurosci ; 34(38): 12893-903, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25232124

ABSTRACT

Intracellular accumulation of amyloid-ß (Aß) protein has been proposed as an early event in AD pathogenesis. In patients with mild cognitive impairment, intraneuronal Aß immunoreactivity was found especially in brain regions critically involved in the cognitive deficits of AD. Although a large body of evidence demonstrates that Aß42 accumulates intraneuronally ((in)Aß), the action and the role of Aß42 buildup on synaptic function have been poorly investigated. Here, we demonstrate that basal synaptic transmission and LTP were markedly depressed following Aß42 injection into the neuron through the patch pipette. Control experiments performed with the reverse peptide (Aß42-1) allowed us to exclude that the effects of (in)Aß depended on changes in oncotic pressure. To further investigate (in)Aß synaptotoxicity we used an Aß variant harboring oxidized methionine in position 35 that does not cross the neuronal plasma membrane and is not uploaded from the extracellular space. This Aß42 variant had no effects on synaptic transmission and plasticity when applied extracellularly, but induced synaptic depression and LTP inhibition after patch-pipette dialysis. Finally, the injection of an antibody raised against human Aß42 (6E10) in CA1 pyramidal neurons of mouse hippocampal brain slices and autaptic microcultures did not, per se, significantly affect LTP and basal synaptic transmission, but it protected against the toxic effects of extracellular Aß42. Collectively, these findings suggest that Aß42-induced impairment of glutamatergic synaptic function depends on its internalization and intracellular accumulation thus paving the way to a systemic proteomic analysis of intracellular targets/partners of Aß42.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Glutamic Acid/physiology , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Synaptic Transmission/drug effects , Amyloid beta-Peptides/administration & dosage , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiology , Intracellular Space/drug effects , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Mice , Microinjections , Neuronal Plasticity/physiology , Peptide Fragments/administration & dosage , Primary Cell Culture , Synaptic Transmission/physiology
9.
Ann Neurol ; 75(4): 602-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24591104

ABSTRACT

Cyclic adenosine monophosphate (cAMP) regulates long-term potentiation (LTP) and ameliorates memory in healthy and diseased brain. Increasing evidence shows that, under physiological conditions, low concentrations of amyloid ß (Aß) are necessary for LTP expression and memory formation. Here, we report that cAMP controls amyloid precursor protein (APP) translation and Aß levels, and that the modulatory effects of cAMP on LTP occur through the stimulation of APP synthesis and Aß production.


Subject(s)
Amyloid beta-Peptides/metabolism , Cyclic AMP/pharmacology , Memory/physiology , Neurons/drug effects , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Hippocampus/cytology , Humans , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Rats , Rats, Sprague-Dawley
10.
J Alzheimers Dis ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38943395

ABSTRACT

This commentary critically examines the long-standing emphasis on amyloid-ß (Aß)-based therapies in Alzheimer's disease (AD), despite numerous clinical trial failures. It highlights the urgency to reassess research methodologies and challenges the initiation of anti-Aß trials in preclinical stages of the disease without conclusive proofs of their safety and efficacy. Instead, a comprehensive approach that considers Aß's physiological roles and addresses AD complex nature is suggested, encouraging the idea that clinical trial failures may result from targeting the wrong mechanism. Evidence-based scientific research is needed to advance with AD treatment, moving beyond the current conception of Aß hypothesis.

11.
Hippocampus ; 23(12): 1367-82, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23939883

ABSTRACT

F3/contactin, a cell-adhesion molecule belonging to the immunoglobulin supergene family, is involved in several aspects of neural development including synapse building, maintenance and functioning. Here, we examine F3/contactin function in adult hippocampal neurogenesis, synaptic plasticity, and memory, using as a model TAG/F3 transgenic mice, where F3/contactin overexpression was induced under control of regulatory sequences from the human TAG-1 (TAX-1) gene. Transgenic mice aged 5 (M5) and 12 (M12) months exhibited an increase in hippocampal size, which correlated with positive effects on precursor proliferation and NeuN expression, these data suggesting a possible role for F3/contactin in promoting adult hippocampal neurogenesis. On the functional level, TAG/F3 mice exhibited increased CA1 long-term potentiation and improved spatial and object recognition memory, notably at 12 months of age. Interestingly, these mice showed an increased expression of the phosphorylated transcription factor CREB, which may represent the main molecular correlate of the observed morphological and functional effects. Altogether, these findings indicate for the first time that F3/contactin plays a role in promoting adult hippocampal neurogenesis and that this effect correlates with improved synaptic function and memory.


Subject(s)
Contactin 1/metabolism , Hippocampus/cytology , Long-Term Potentiation/genetics , Memory/physiology , Neurogenesis/genetics , Age Factors , Animals , Bromodeoxyuridine/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cell Proliferation , Contactin 1/genetics , Electric Stimulation , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/physiology , In Vitro Techniques , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques , Recognition, Psychology/physiology
12.
Ann Neurol ; 69(5): 819-30, 2011 May.
Article in English | MEDLINE | ID: mdl-21472769

ABSTRACT

OBJECTIVE: The goal of this study was to investigate the role of endogenous amyloid-ß peptide (Aß) in healthy brain. METHODS: Long-term potentiation (LTP), a type of synaptic plasticity that is thought to be associated with learning and memory, was examined through extracellular field recordings from the CA1 region of hippocampal slices, whereas behavioral techniques were used to assess contextual fear memory and reference memory. Amyloid precursor protein (APP) expression was reduced through small interfering RNA (siRNA) technique. RESULTS: We found that both antirodent Aß antibody and siRNA against murine APP reduced LTP as well as contextual fear memory and reference memory. These effects were rescued by the addition of human Aß42, suggesting that endogenously produced Aß is needed for normal LTP and memory. Furthermore, the effect of endogenous Aß on plasticity and memory was likely due to regulation of transmitter release, activation of α7-containing nicotinic acetylcholine receptors, and Aß42 production. INTERPRETATION: Endogenous Aß42 is a critical player in synaptic plasticity and memory within the normal central nervous system. This needs to be taken into consideration when designing therapies aiming at reducing Aß levels to treat Alzheimer disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Memory/physiology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Antibodies/pharmacology , Behavior, Animal/drug effects , Biophysics/methods , Electric Stimulation/methods , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/physiology , Hippocampus/drug effects , Humans , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Inbred C57BL , Peptide Fragments/metabolism , RNA, Small Interfering/pharmacology
13.
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
14.
J Alzheimers Dis ; 85(3): 1343-1356, 2022.
Article in English | MEDLINE | ID: mdl-34924388

ABSTRACT

BACKGROUND: Object recognition task (ORT) is a widely used behavioral paradigm to assess memory in rodent models, due to its easy technical execution, the lack of aversive stressful stimuli, and the possibility to repeat the test on the same animals. However, mouse exploration might be strongly influenced by a variety of variables. OBJECTIVE: To study whether innate preferences influenced exploration in male and female wild type mice and the Alzheimer's disease (AD) model 3xTg. METHODS: We first evaluated how object characteristics (material, size, and shape) influence exploration levels, latency, and exploration modality. Based on these findings, we evaluated whether these innate preferences biased the results of ORT performed in wild type mice and AD models. RESULTS: Assessment of Exploration levels, i.e., the time spent in exploring a certain object in respect to the total exploration time, revealed an innate preference for objects made in shiny materials, such as metal and glass. A preference for bigger objects characterized by higher affordance was also evident, especially in male mice. When performing ORT, exploration was highly influenced by these innate preferences. Indeed, both wild type and AD mice spent more time in exploring the metal object, regardless of its novelty. Furthermore, the use of objects with higher affordance such as the cube was a confounding factor leading to "false" results that distorted ORT interpretation. CONCLUSION: When designing exploration-based behavioral experiments aimed at assessing memory in healthy and AD mice, object characteristics should be carefully evaluated to improve scientific outcomes and minimize possible biases.


Subject(s)
Alzheimer Disease/psychology , Exploratory Behavior/physiology , Object Attachment , Visual Perception/physiology , Animals , Disease Models, Animal , Female , Male , Mice
15.
eNeuro ; 9(5)2022.
Article in English | MEDLINE | ID: mdl-36104275

ABSTRACT

Implantation of guide cannulas is a widely used technique to access specific brain areas. Although commercially available, the need to personalize these implants and the high cost prompted us to design open-source customized devices taking advantage of 3D printing technology. Our cannulas consisted in a 3D-printed head mount designed according to the Paxinos coordinates to reach the CA1 area of the hippocampus. To cut guide cannulas to the proper length, we designed and realized an original 3D-printed linear motion apparatus. Polylactic acid thermoplastic polymer was used as printing material. Homemade or commercial cannulas were implanted in 4- to 6-month-old wild-type mice and intrahippocampal injections of amyloid-ß peptide at different concentrations were performed. In vivo behavioral studies of novel object recognition indicated that results obtained with homemade versus commercial devices were comparable. Methylene blue injections and Nissl staining confirmed the correct localization of cannulas in the CA1 area of mouse hippocampus. Our method allows a fast manufacturing of hippocampal cannulas preserving the required precision at very low cost. Furthermore, this system can be easily modified to produce cannulas to target other brain areas. In conclusion, 3D printing might be used as a useful and versatile technology to realize open-source customized devices in neuroscience laboratories.


Subject(s)
Cannula , Methylene Blue , Animals , Hippocampus , Mice , Peptides , Polymers , Printing, Three-Dimensional
16.
Neuron ; 56(4): 670-88, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18031684

ABSTRACT

Trafficking of AMPA receptors (AMPARs) is regulated by specific interactions of the subunit intracellular C-terminal domains (CTDs) with other proteins, but the mechanisms involved in this process are still unclear. We have found that the GluR1 CTD binds to cGMP-dependent protein kinase II (cGKII) adjacent to the kinase catalytic site. Binding of GluR1 is increased when cGKII is activated by cGMP. cGKII and GluR1 form a complex in the brain, and cGKII in this complex phosphorylates GluR1 at S845, a site also phosphorylated by PKA. Activation of cGKII by cGMP increases the surface expression of AMPARs at extrasynaptic sites. Inhibition of cGKII activity blocks the surface increase of GluR1 during chemLTP and reduces LTP in the hippocampal slice. This work identifies a pathway, downstream from the NMDA receptor (NMDAR) and nitric oxide (NO), which stimulates GluR1 accumulation in the plasma membrane and plays an important role in synaptic plasticity.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Hippocampus/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Receptors, AMPA/metabolism , Synaptic Transmission/physiology , Animals , Catalytic Domain , Cell Line , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type II , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hippocampus/ultrastructure , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/ultrastructure , Organ Culture Techniques , Phosphorylation , Protein Binding/physiology , Protein Transport/drug effects , Protein Transport/physiology , Rats , Signal Transduction/drug effects , Signal Transduction/physiology
17.
Front Mol Neurosci ; 14: 684977, 2021.
Article in English | MEDLINE | ID: mdl-34211368

ABSTRACT

The main goal of scientific research is to uncover new knowledge to understand reality. In the field of life sciences, the aim of translational research-to transfer results "from bench to bedside"-has to contend with the problem that the knowledge acquired at the "bench" is often not reproducible at the "bedside," raising the question whether scientific discoveries truly mirror the real world. As a result, researchers constantly struggle to overcome the dichotomy between methodological problems and expectations, as funding agencies and industries demand expandable and quick results whereas patients, who are uninterested in the epistemological dispute, only ask for an effective cure. Despite the numerous attempts made to address reproducibility and reliability issues, some essential pitfalls of scientific investigations are often overlooked. Here, we discuss some limitations of the conventional scientific method and how researcher cognitive bias and conceptual errors have the potential to steer an experimental study away from the search for the vera causa of a phenomenon. As an example, we focus on Alzheimer's disease research and on some problems that may have undermined most of the clinical trials conducted to investigate it.

18.
Prog Neurobiol ; 206: 102154, 2021 11.
Article in English | MEDLINE | ID: mdl-34453977

ABSTRACT

The accumulation of amyloid-beta peptide (Aß) and the failure of cholinergic transmission are key players in Alzheimer's disease (AD). However, in the healthy brain, Aß contributes to synaptic plasticity and memory acting through α7 subtype nicotinic acetylcholine receptors (α7nAChRs). Here, we hypothesized that the α7nAChR deletion blocks Aß physiological function and promotes a compensatory increase in Aß levels that, in turn, triggers an AD-like pathology. To validate this hypothesis, we studied the age-dependent phenotype of α7 knock out mice. We found that α7nAChR deletion caused an impairment of hippocampal synaptic plasticity and memory at 12 months of age, paralleled by an increase of Amyloid Precursor Protein expression and Aß levels. This was accompanied by other classical AD features such as a hyperphosphorylation of tau at residues Ser 199, Ser 396, Thr 205, a decrease of GSK-3ß at Ser 9, the presence of paired helical filaments and neurofibrillary tangles, neuronal loss and an increase of GFAP-positive astrocytes. Our findings suggest that α7nAChR malfunction might precede Aß and tau pathology, offering a different perspective to interpret the failure of anti-Aß therapies against AD and to find novel therapeutical approaches aimed at restoring α7nAChRs-mediated Aß function at the synapse.


Subject(s)
Alzheimer Disease , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Glycogen Synthase Kinase 3 beta , Mice , Peptide Fragments/metabolism , Receptors, Nicotinic/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics
19.
Front Pharmacol ; 12: 809541, 2021.
Article in English | MEDLINE | ID: mdl-35002742

ABSTRACT

Depression is a risk factor for the development of Alzheimer's disease (AD). A neurobiological and clinical continuum exists between AD and depression, with neuroinflammation and oxidative stress being involved in both diseases. Second-generation antidepressants, in particular selective serotonin reuptake inhibitors (SSRIs), are currently investigated as neuroprotective drugs in AD. By employing a non-transgenic AD model, obtained by intracerebroventricular (i.c.v.) injection of amyloid-ß (Aß) oligomers in 2-month-old C57BL/6 mice, we recently demonstrated that the SSRI fluoxetine (FLX) and the multimodal antidepressant vortioxetine (VTX) reversed the depressive-like phenotype and memory deficits induced by Aß oligomers rescuing the levels of transforming growth factor-ß1 (TGF-ß1). Aim of our study was to test FLX and VTX for their ability to prevent oxidative stress in the hippocampus of Aß-injected mice, a brain area strongly affected in both depression and AD. The long-term intraperitoneal (i.p.) administration of FLX (10 mg/kg) or VTX (5 and 10 mg/kg) for 24 days, starting 7 days before Aß injection, was able to prevent the over-expression of inducible nitric oxide synthase (iNOS) and NADPH oxidase 2 (Nox2) induced by Aß oligomers. Antidepressant pre-treatment was also able to rescue the mRNA expression of glutathione peroxidase 1 (Gpx1) antioxidant enzyme. FLX and VTX also prevented Aß-induced neurodegeneration in mixed neuronal cultures treated with Aß oligomers. Our data represent the first evidence that the long-term treatment with the antidepressants FLX or VTX can prevent the oxidative stress phenomena related to the cognitive deficits and depressive-like phenotype observed in a non-transgenic animal model of AD.

20.
Neurobiol Stress ; 14: 100286, 2021 May.
Article in English | MEDLINE | ID: mdl-33392367

ABSTRACT

Translational animal models for studying post-traumatic stress disorder (PTSD) are valuable for elucidating the poorly understood neurobiology of this neuropsychiatric disorder. These models should encompass crucial features, including persistence of PTSD-like phenotypes triggered after exposure to a single traumatic event, trauma susceptibility/resilience and predictive validity. Here we propose a novel arousal-based individual screening (AIS) model that recapitulates all these features. The AIS model was designed by coupling the traumatization (24 h restraint) of C57BL/6 J mice with a novel individual screening. This screening consists of z-normalization of post-trauma changes in startle reactivity, which is a measure of arousal depending on neural circuits conserved across mammals. Through the AIS model, we identified susceptible mice showing long-lasting hyperarousal (up to 56 days post-trauma), and resilient mice showing normal arousal. Susceptible mice further showed persistent PTSD-like phenotypes including exaggerated fear reactivity and avoidance of trauma-related cue (up to 75 days post-trauma), increased avoidance-like behavior and social/cognitive impairment. Conversely, resilient mice adopted active coping strategies, behaving like control mice. We further uncovered novel transcriptional signatures driven by PTSD-related genes as well as dysfunction of hypothalamic-pituitary-adrenal axis, which corroborated the segregation in susceptible/resilient subpopulations obtained through the AIS model and correlated with trauma susceptibility/resilience. Impaired hippocampal synaptic plasticity was also observed in susceptible mice. Finally, chronic treatment with paroxetine ameliorated the PTSD-like phenotypes of susceptible mice. These findings indicate that the AIS model might be a new translational animal model for the study of crucial features of PTSD. It might shed light on the unclear PTSD neurobiology and identify new pharmacological targets for this difficult-to-treat disorder.

SELECTION OF CITATIONS
SEARCH DETAIL