Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
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.
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
3.
Eur J Neurosci ; 39(6): 893-903, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24382162

ABSTRACT

In recent years, much effort has been devoted to identifying stimuli capable of enhancing adult neurogenesis, a process that generates new neurons throughout life, and that appears to be dysfunctional in the senescent brain and in several neuropsychiatric and neurodegenerative diseases. We previously reported that in vivo exposure to extremely low-frequency electromagnetic fields (ELFEFs) promotes the proliferation and neuronal differentiation of hippocampal neural stem cells (NSCs) that functionally integrate in the dentate gyrus. Here, we extended our studies to specifically assess the influence of ELFEFs on hippocampal newborn cell survival, which is a very critical issue in adult neurogenesis regulation. Mice were injected with 5-bromo-2'-deoxyuridine (BrdU) to label newborn cells, and were exposed to ELFEFs 9 days later, when the most dramatic decrease in the number of newly generated neurons occurs. The results showed that ELFEF exposure (3.5 h/day for 6 days) enhanced newborn neuron survival as documented by double staining for BrdU and doublecortin, to identify immature neurons, or NeuN labeling of mature neurons. The effects of ELFEFs were associated with enhanced spatial learning and memory. In an in vitro model of hippocampal NSCs, ELFEFs exerted their pro-survival action by rescuing differentiating neurons from apoptotic cell death. Western immunoblot assay revealed reduced expression of the pro-apoptotic protein Bax, and increased levels of the anti-apoptotic protein Bcl-2, in the hippocampi of ELFEF-exposed mice as well as in ELFEF-exposed NSC cultures, as compared with their sham-exposed counterparts. Our results may have clinical implications for the treatment of impaired neurogenesis associated with brain aging and neurodegenerative diseases.


Subject(s)
Apoptosis , Electromagnetic Fields , Hippocampus/radiation effects , Neurons/radiation effects , Animals , Cell Survival/radiation effects , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/physiology , Male , Maze Learning , Memory , Mice , Mice, Inbred C57BL , Neurogenesis , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
4.
Brain ; 135(Pt 12): 3735-48, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23183235

ABSTRACT

Alzheimer's disease is a devastating cureless neurodegenerative disorder affecting >35 million people worldwide. The disease is caused by toxic oligomers and aggregates of amyloid ß protein and the microtubule-associated protein tau. Recently, the Lys-specific molecular tweezer CLR01 has been shown to inhibit aggregation and toxicity of multiple amyloidogenic proteins, including amyloid ß protein and tau, by disrupting key interactions involved in the assembly process. Following up on these encouraging findings, here, we asked whether CLR01 could protect primary neurons from Alzheimer's disease-associated synaptotoxicity and reduce Alzheimer's disease-like pathology in vivo. Using cell culture and brain slices, we found that CLR01 effectively inhibited synaptotoxicity induced by the 42-residue isoform of amyloid ß protein, including ∼80% inhibition of changes in dendritic spines density and long-term potentiation and complete inhibition of changes in basal synaptic activity. Using a radiolabelled version of the compound, we found that CLR01 crossed the mouse blood-brain barrier at ∼2% of blood levels. Treatment of 15-month-old triple-transgenic mice for 1 month with CLR01 resulted in a decrease in brain amyloid ß protein aggregates, hyperphosphorylated tau and microglia load as observed by immunohistochemistry. Importantly, no signs of toxicity were observed in the treated mice, and CLR01 treatment did not affect the amyloidogenic processing of amyloid ß protein precursor. Examining induction or inhibition of the cytochrome P450 metabolism system by CLR01 revealed minimal interaction. Together, these data suggest that CLR01 is safe for use at concentrations well above those showing efficacy in mice. The efficacy and toxicity results support a process-specific mechanism of action of molecular tweezers and suggest that these are promising compounds for developing disease-modifying therapy for Alzheimer's disease and related disorders.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Brain/pathology , Lysine/chemistry , Neurons/physiology , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Animals , Antiparasitic Agents/chemistry , Antiparasitic Agents/therapeutic use , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/physiology , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Electric Stimulation , Exploratory Behavior/drug effects , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Lysine/pharmacology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Protein Isoforms/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , tau Proteins/genetics
5.
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
6.
J Clin Invest ; 130(9): 4831-4844, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32544084

ABSTRACT

The amyloid hypothesis posits that the amyloid-beta (Aß) protein precedes and requires microtubule-associated protein tau in a sort of trigger-bullet mechanism leading to Alzheimer's disease (AD) pathology. This sequence of events has become dogmatic in the AD field and is used to explain clinical trial failures due to a late start of the intervention when Aß already activated tau. Here, using a multidisciplinary approach combining molecular biological, biochemical, histopathological, electrophysiological, and behavioral methods, we demonstrated that tau suppression did not protect against Aß-induced damage of long-term synaptic plasticity and memory, or from amyloid deposition. Tau suppression could even unravel a defect in basal synaptic transmission in a mouse model of amyloid deposition. Similarly, tau suppression did not protect against exogenous oligomeric tau-induced impairment of long-term synaptic plasticity and memory. The protective effect of tau suppression was, in turn, confined to short-term plasticity and memory. Taken together, our data suggest that therapies downstream of Aß and tau together are more suitable to combat AD than therapies against one or the other alone.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Long-Term Potentiation , Synapses/metabolism , Synaptic Transmission , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Mice , Mice, Knockout , Synapses/genetics , Synapses/pathology , tau Proteins/genetics
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.
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
9.
Front Pharmacol ; 5: 97, 2014.
Article in English | MEDLINE | ID: mdl-24847267

ABSTRACT

Among the multiple factors concurring to Alzheimer's disease (AD) pathogenesis, greater attention should be devoted to the role played by infectious agents. Growing epidemiological and experimental evidence suggests that recurrent herpes simplex virus type-1 (HSV-1) infection is a risk factor for AD although the underlying molecular and functional mechanisms have not been fully elucidated yet. Here, we review literature suggesting the involvement of HSV-1 infection in AD also briefly mentioning possible pharmacological implications of these findings.

10.
Neurobiol Aging ; 34(4): 1032-44, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23046860

ABSTRACT

Perisynaptic accumulations of amyloid ß-protein (Aß) play a critical role in the synaptic dysfunction underlying the cognitive impairment observed in Alzheimer's disease. The methionine residue at position 35 (Met35) in Aß is highly subject to oxidation in Alzheimer's disease brains. In hippocampal brain slices we found that long-term potentiation at CA3-CA1 synapses was significantly inhibited by wild type Aß42 in which Met35 is reduced, but not by Aß42 harboring Met35 sulfoxide. Similar differences were observed when basal synaptic transmission was investigated in autaptic hippocampal neurons. The significant decreases in excitatory postsynaptic current amplitude, vesicle release probability and miniature excitatory postsynaptic current frequency caused by 20-minute exposure to wild type Aß42 were not observed after exposure to Aß42 harboring Met35 sulfoxide. With longer (24-hour) Aß treatments, this early impairment of the presynaptic terminal function extended to involve the postsynaptic side as well. The Met35 oxidation also affected Aß42 negative impact on dendritic spine density and expression of pre- and postsynaptic proteins (synaptophysin and postsynaptic density protein-95). Our findings suggest that oxidation of Met35 is critical for molecular, structural, and functional determinants of Aß42 synaptotoxicity.


Subject(s)
Action Potentials/physiology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/pharmacology , Neurons/physiology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Synapses/physiology , Synaptic Transmission/physiology , Action Potentials/drug effects , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Structure-Activity Relationship , Synapses/drug effects , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL