Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
PLoS One ; 17(4): e0267164, 2022.
Article in English | MEDLINE | ID: mdl-35421203

ABSTRACT

Extracellular vesicles (EVs) are double membrane structures released by all cell types with identified roles in the generation, transportation, and degradation of amyloid-ß protein (Aß) oligomers in Alzheimer's disease (AD). EVs are thus increasingly recognized to play a neuroprotective role in AD, through their ability to counteract the neurotoxic effects of Aß, possibly through interactions with specific receptors on cell membranes. Our previous studies have identified the amylin receptor (AMY), particularly AMY3 subtype, as a mediator of the deleterious actions of Aß in vitro and in vivo experimental paradigms. In the present study, we demonstrate that AMY3 enriched EVs can bind soluble oligomers of Aß and protect N2a cells against toxic effects of this peptide. The effect was specific to amylin receptor as it was blocked in the presence of amylin receptor antagonist AC253. This notion was supported by reduced Aß binding to EVs from AMY depleted mice compared to those from wild type (Wt) mice. Finally, application of AMY3, but not Wt derived, EVs to hippocampal brain slices improved Aß-induced reduction of long-term potentiation, a cellular surrogate of memory. Collectively, our observations support the role of AMY receptors, particularly AMY3, in EVs as a potential therapeutic target for AD.


Subject(s)
Alzheimer Disease , Extracellular Vesicles , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Extracellular Vesicles/metabolism , Long-Term Potentiation , Mice , Peptide Fragments/toxicity , Receptors, Islet Amyloid Polypeptide/metabolism
3.
Mol Neurobiol ; 58(10): 5369-5382, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34312771

ABSTRACT

Based upon its interactions with amyloid ß peptide (Aß), the amylin receptor, a class B G protein-coupled receptor (GPCR), is a potential modulator of Alzheimer's disease (AD) pathogenesis. However, past pharmacological approaches have failed to resolve whether activation or blockade of this receptor would have greater therapeutic benefit. To address this issue, we generated compound mice expressing a human amyloid precursor protein gene with familial AD mutations in combination with deficiency of amylin receptors produced by hemizygosity for the critical calcitonin receptor subunit of this heterodimeric GPCR. These compound transgenic AD mice demonstrated attenuated responses to human amylin- and Aß-induced depression of hippocampal long-term potentiation (LTP) in keeping with the genetic depletion of amylin receptors. Both the LTP responses and spatial memory (as measured with Morris water maze) in these mice were improved compared to AD mouse controls and, importantly, a reduction in both the amyloid plaque burden and markers of neuroinflammation was observed. Our data support the notion of further development of antagonists of the amylin receptor as AD-modifying therapies.


Subject(s)
Alzheimer Disease/genetics , Maze Learning/physiology , Receptors, Calcitonin/genetics , Receptors, Islet Amyloid Polypeptide/genetics , Spatial Memory/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , Endothelium, Vascular/metabolism , Excitatory Postsynaptic Potentials/physiology , Female , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Receptors, Calcitonin/deficiency , Receptors, Islet Amyloid Polypeptide/deficiency
4.
Sci Rep ; 9(1): 10942, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358858

ABSTRACT

Recent evidence supports involvement of amylin and the amylin receptor in the pathogenesis of Alzheimer's disease (AD). We have previously shown that amylin receptor antagonist, AC253, improves spatial memory in AD mouse models. Herein, we generated and screened a peptide library and identified two short sequence amylin peptides (12-14 aa) that are proteolytically stable, brain penetrant when administered intraperitoneally, neuroprotective against Aß toxicity and restore diminished levels of hippocampal long term potentiation in AD mice. Systemic administration of the peptides for five weeks in aged 5XFAD mice improved spatial memory, reduced amyloid plaque burden, and neuroinflammation. The common residue SQELHRLQTY within the peptides is an essential sequence for preservation of the beneficial effects of the fragments that we report here and constitutes a new pharmacological target. These findings suggest that the amylin receptor antagonism may represent a novel therapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Animals , Female , Hippocampus/drug effects , Islet Amyloid Polypeptide/chemistry , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Receptors, Islet Amyloid Polypeptide/metabolism , Spatial Memory
5.
Trends Mol Med ; 23(8): 709-720, 2017 08.
Article in English | MEDLINE | ID: mdl-28694141

ABSTRACT

Alzheimer'sdisease (AD) is a progressive neurodegenerative disorder, characterized by senile plaques constituting extracellular deposits of ß-amyloid (Aß) fibrils. Since Aß accumulation in the brain is considered an early event preceding, by decades, cognitive dysfunction, disease-modifying treatments are aimed at facilitating clearance of this protein from the brain or ameliorating its toxic effects. Recent studies have identified the amylin receptor as a capable mediator of the deleterious actions of Aß and furthermore, administration of amylin receptor-based peptides has been shown to improve spatial memory and learning in transgenic mouse models of AD. Here, by discussing available evidence, we posit that the amylin receptor could be considered a potential therapeutic target for AD, and present the rationale for using amylin receptor antagonists to treat this debilitating condition.


Subject(s)
Alzheimer Disease/drug therapy , Peptides/therapeutic use , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Humans , Mice , Mice, Transgenic , Receptors, Islet Amyloid Polypeptide/genetics
6.
J Neurosci ; 32(48): 17401-6, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197731

ABSTRACT

Alzheimer's disease (AD) is characterized by accumulation of amyloid-ß peptide (Aß) in the brain regions that subserve memory and cognition. The amylin receptor is a potential target receptor for expression of the deleterious actions of soluble oligomeric Aß species. We investigated whether the amylin receptor antagonist, AC253, neutralizes the depressant effects of Aß(1-42) and human amylin on hippocampal long-term potentiation (LTP). Furthermore, we examined whether depressed levels of LTP observed in transgenic mice, which overexpress amyloid precursor protein (TgCRND8), could be restored with AC253. In mouse hippocampal brain slices, field EPSPs were recorded from the stratum radiatum layer of the CA1 area (cornu ammonis 1 region of the hippocampus) in response to electrical stimulation of Schaeffer collateral afferents. LTP was induced by 3-theta burst stimulation protocols. Aß(1-42) (50 nM) and human amylin (50 nM), but not Aß(42-1) (50 nM), depressed LTP evoked using both stimulation protocols. Preapplication of AC253 (250 nM) blocked Aß- and human amylin-induced reduction of LTP without affecting baseline transmission or LTP on its own. In contrast to wild-type controls, where robust LTP is observed, 6- to 12-month-old TgCRND8 mice show blunted LTP that is significantly enhanced by application of AC253. Our data demonstrate that the effects of Aß(1-42) and human amylin on LTP are expressed via the amylin receptor, and moreover, blockade of this receptor increases LTP in transgenic mice that show increased brain amyloid burden. Amylin receptor antagonists could serve as potentially useful therapeutic agents in AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Hippocampus/drug effects , Islet Amyloid Polypeptide/pharmacology , Long-Term Potentiation/drug effects , Peptide Fragments/pharmacology , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Amyloid beta-Protein Precursor/genetics , Animals , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/physiology , Long-Term Potentiation/physiology , Male , Mice , Mice, Transgenic
7.
CNS Neurosci Ther ; 18(8): 641-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22613619

ABSTRACT

AIMS: To evaluate the acute effects of the mitochondrial complex I inhibitor rotenone on rat hippocampal synaptic plasticity. METHODS: Electrophysiological field potential recordings were used to measure basal synaptic transmission and synaptic plasticity in rat coronal hippocampal slices. Synaptic long-term potentiation (LTP) was induced by high-frequency stimulation (100 Hz, 1 second × 3 at an interval of 20 seconds). In addition, mitochondrial complex I function was measured using MitoSOX imaging in mitochondrial preparations. RESULTS: Acute exposure of hippocampal slices to 50 nM rotenone for 1 h did not alter basal CA3-CA1 synaptic transmission though 500 nM rotenone significantly reduced basal synaptic transmission. However, 50 nM rotenone significantly impaired LTP and this rotenone's effect was prevented by co-application of rotenone plus the ketones acetoacetate and ß-hydroxybutyrate (1 mM each). Finally, we measured mitochondrial function using MitoSOX imaging in mitochondrial preparations and found that 50 nM rotenone partially reduced mitochondrial function whereas 500 nM rotenone completely eliminated mitochondrial function. CONCLUSIONS: Our findings suggest that mitochondrial activity driven by complex I is a sensitive modulator of synaptic plasticity in the hippocampus. Acute exposure of the hippocampus to rotenone eliminates complex I function and in turn impairs LTP.


Subject(s)
Electron Transport Complex I/drug effects , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Rotenone/toxicity , Synapses/drug effects , Uncoupling Agents/toxicity , 3-Hydroxybutyric Acid/pharmacology , Acetoacetates/pharmacology , Animals , CA1 Region, Hippocampal/drug effects , Electrophysiological Phenomena , In Vitro Techniques , Ketones/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Neuronal Plasticity/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Synaptic Transmission/drug effects
8.
J Neurochem ; 113(1): 248-61, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20089133

ABSTRACT

beta-Site amyloid precursor protein cleaving enzyme 1 (BACE1) initiates amyloid-beta (Abeta) generation that is central to the pathophysiology of Alzheimer's disease (AD). Therefore, lowering Abeta levels by BACE1 manipulations represents a key therapeutic strategy, but it remains unclear whether partial inhibition of BACE1, as expected for AD treatments, can improve memory deficits. In this study, we used heterozygous BACE1 gene knockout (BACE1+/-) mice to evaluate the effects of partial BACE1 suppression on different types of synaptic and cognitive dysfunctions in Alzheimer's transgenic mice (5XFAD model). We found that approximately 50% BACE1 reductions rescued deficits of 5XFAD mice not only in hippocampus-dependent memories as tested by contextual fear conditioning and spontaneous alternation Y-maze paradigms but also in cortex-dependent remote memory stabilization during 30 days after contextual conditioning. Furthermore, 5XFAD-associated impairments in long-term potentiation (a synaptic model of learning and memory) and declines in synaptic plasticity/learning-related brain-derived neurotrophic factor-tyrosine kinase B signaling pathways were prevented in BACE1+/-.5XFAD mice. Finally, these improvements were related with reduced levels of beta-secretase-cleaved C-terminal fragment (C99), Abeta peptides and plaque burden in relevant brain regions of BACE1+/-.5XFAD mice. Therefore, our findings provide compelling evidence for beneficial effects of partially BACE1-inhibiting approaches on multiple forms of functional defects associated with AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Memory Disorders/etiology , Neuronal Plasticity/genetics , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Conditioning, Classical/physiology , Disease Models, Animal , Electric Stimulation/methods , Enzyme-Linked Immunosorbent Assay/methods , Excitatory Postsynaptic Potentials/genetics , Fear/physiology , Gene Expression Regulation/genetics , Humans , In Vitro Techniques , Long-Term Potentiation/genetics , Maze Learning/physiology , Mice , Mice, Transgenic , Mutation/genetics , Neuronal Plasticity/drug effects , Patch-Clamp Techniques/methods , Peptide Fragments/metabolism , Presenilin-1/genetics
9.
Neurobiol Dis ; 33(2): 229-35, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19026746

ABSTRACT

Although animal models of Alzheimer's disease (AD) recapitulate beta-amyloid-dependent hippocampal synaptic and cognitive dysfunctions, it is poorly understood how cortex-dependent remote memory stabilization following initial hippocampal coding is affected. Here, we systematically analyzed biophysical and behavioral phenotypes, including remote memory functions, of 5XFAD APP/PS1 transgenic mice containing five familial AD mutations. We found that 5XFAD mice show hippocampal dysfunctions as observed by reduced levels of baseline transmission and long-term potentiation at Schaffer collateral-CA1 synapses. Hippocampus-dependent memory tested 1 day after contextual fear conditioning was also impaired age-dependently in 5XFAD mice, as correlated with the onset of hippocampal synaptic failures. Importantly, remote memory stabilization during 30 days after training significantly declined in 5XFAD mice at time well before the onset of hippocampal dysfunctions. Our results indicate that 5XFAD mice provide a useful model system to investigate the mechanisms and therapeutic interventions for multiple synaptic and memory dysfunctions associated with AD.


Subject(s)
Alzheimer Disease/physiopathology , Disease Models, Animal , Hippocampus/physiopathology , Long-Term Potentiation , Memory/physiology , Synaptic Transmission , Age Factors , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Animals , Conditioning, Psychological , Excitatory Postsynaptic Potentials , Extinction, Psychological/physiology , Fear/psychology , Humans , Mice , Mice, Transgenic , Mutation , Presenilin-1/genetics , Protease Nexins , Receptors, Cell Surface/genetics
10.
Learn Mem ; 15(11): 837-43, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18984565

ABSTRACT

Accumulating evidence indicates the key role of alpha-calcium/calmodulin-dependent protein kinase II (alphaCaMKII) in synaptic plasticity and learning, but it remains unclear how this kinase participates in the processing of memory extinction. Here, we investigated the mechanism by which alphaCaMKII may mediate extinction by using heterozygous knock-in mice with a targeted T286A mutation that prevents the autophosphorylation of this kinase (alphaCaMKII(T286A+/-)). Remarkably, partial reduction of alphaCaMKII function due to the T286A(+/-) mutation prevented the development of extinction without interfering with initial hippocampus-dependent memory formation as assessed by contextual fear conditioning and the Morris water maze. It is hypothesized that the mechanism of extinction may differ depending on the interval at which extinction training is started, being more akin to "new learning" at longer intervals and "unlearning" or "erasure" at shorter intervals. Consistent with this hypothesis, we found that extinction conducted 24 h, but not 15 min, after contextual fear training showed spontaneous recovery (reappearance of extinguished freezing responses) 21 d following the extinction, representing behavioral evidence for new learning and unlearning mechanisms underlying extinction 24 h and 15 min post-training, respectively. Importantly, the alphaCaMKII(T286A+/-) mutation blocked new learning of contextual fear memory extinction, whereas it did not interfere with unlearning processes. Our results demonstrate a genetic dissociation of new learning and unlearning mechanisms of extinction, and suggest that alphaCaMKII is responsible for extinguishing memories specifically through new learning mechanisms.


Subject(s)
Association Learning/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Conditioning, Classical/physiology , Extinction, Psychological/physiology , Memory Disorders/enzymology , Animals , Behavior, Animal , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cues , Fear , Freezing Reaction, Cataleptic/physiology , Maze Learning/physiology , Memory Disorders/genetics , Mice , Mice, Mutant Strains , Mutation , Phosphorylation , Space Perception/physiology
11.
Peptides ; 27(7): 1814-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16513211

ABSTRACT

The actions of the corticotropin-releasing factor (CRF) family of peptides are mediated by the seven transmembrane-domain G-protein-coupled receptors, the CRF receptors type 1 (CRF1 receptor) and type 2 (CRF2 receptor). In a previous study, we reported that CRF, an endogenous ligand for CRF1 receptor, modulated Ca2+ influx in rat pancreatic beta-cells. In addition to CRF, other additional members of the family, urocortins, have been identified in mammals. Urocortin 1 (UCN 1), a peptide of the CRF family, binds both CRF1 receptor and CRF2 receptor with equal affinities. Urocortin 3 (UCN 3), a highly selective ligand for CRF2 receptor with little affinity for CRF1 receptor, has been shown in rat pancreatic beta-cells. The present study focused on the effects of the CRF family peptides on intracellular Ca2+ ([Ca2+]i) concentration via CRF receptors in rat pancreatic beta-cells. Microfluorimetric experiments showed that CRF (0.2 nM) and UCN 1 (0.2 nM) elevated [Ca2+]i levels. Both CRF and UCN 1 effects were attenuated by astressin, a non-selective CRF receptor antagonist. Antisauvagine-30, a selective CRF2 receptor antagonist, appeared to enhance the UCN 1 effect on the elevation of [Ca2+]i. The CRF effect on the elevation of [Ca2+]i was inhibited by the addition of UCN 3. Taken together, the activation of CRF2 receptor antagonizes the CRF1 receptor-stimulated Ca2+ influx.


Subject(s)
Calcium/metabolism , Corticotropin-Releasing Hormone/metabolism , Insulin-Secreting Cells/metabolism , Animals , Corticotropin-Releasing Hormone/pharmacology , Dose-Response Relationship, Drug , Ligands , Peptide Fragments/pharmacology , Peptides/chemistry , Rats , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins
12.
EMBO J ; 25(4): 673-82, 2006 Feb 22.
Article in English | MEDLINE | ID: mdl-16467850

ABSTRACT

Individual exocytic events in intact adrenal medulla were visualized by two-photon extracellular polar-tracer imaging. Exocytosis of chromaffin vesicles often occurred in a sequential manner, involving first vesicles located at the cell periphery and then those present deeper within the cytoplasm. Sequential exocytosis occurred preferentially at regions of the plasma membrane facing the intercellular space. The compound vesicles swelled to more than five times their original volume and formed vacuolar exocytic lumens as a result of expansion of intravesicular gels and their confinement within the lumen by the fusion pore and the narrow intercellular space. Such luminal swelling greatly promoted sequential exocytosis. The SNARE protein SNAP25 rapidly migrated from the plasma membrane to the membrane of fused vesicles. These data indicate that vesicles present deeper within the cytoplasm can be fusion ready like those at the cell periphery, and that swelling of exocytic lumens promotes assembly of the fusion machinery. We suggest the existence of two molecular configurations for fusion-ready states in Ca2+ -dependent exocytosis.


Subject(s)
Adrenal Medulla/physiology , Cell Membrane/metabolism , Chromaffin Cells/physiology , Exocytosis/physiology , Synaptosomal-Associated Protein 25/metabolism , Vacuoles/physiology , Adrenal Medulla/cytology , Animals , Calcium/metabolism , Cattle , Chromaffin Cells/cytology , Microscopy, Confocal , Protein Transport/physiology , Rats
13.
Neuropharmacology ; 48(6): 869-80, 2005 May.
Article in English | MEDLINE | ID: mdl-15829257

ABSTRACT

Hippocampal gamma oscillations, as a form of neuronal network synchronization, are speculated to be associated with learning, memory and attention. Nicotinic acetylcholine receptor alpha7 subtypes (alpha7-nAChRs) are highly expressed in hippocampal neurons and play important roles in modulating neuronal function, synaptic plasticity, learning and memory. However, little is known about the role of alpha7-nAChRs in hippocampal gamma oscillations. Here, we examined the effects of selective alpha7- and non-alpha7-nAChR antagonists on tetanic gamma oscillations in rat hippocampal slices. We found that brief tetanic stimulation-induced gamma oscillations (30-80 Hz) and pharmacological blockade of alpha7-nAChRs using the relatively selective alpha7-nAChR antagonists, methyllycaconitine (10 or 100 nM) or alpha-bungarotoxin (10 nM), significantly reduced the frequency spectrum power, the number of spikes, and burst duration of evoked gamma oscillations. Neither mecamylamine nor dihydro-beta-erythroidine, which are selective antagonists of non-alpha7-nAChRs, demonstrated significant effects on tetanic gamma oscillations. Nicotine exposure promotes hippocampal gamma oscillations in a methyllycaconitine-sensitive manner. It is concluded that alpha7-nAChRs in hippocampal slices play important roles in regulation of gamma oscillations, thus potentially helping to explain roles of nAChRs in cognitive functions such as learning, memory and attention.


Subject(s)
Aconitine/analogs & derivatives , Biological Clocks/radiation effects , Electric Stimulation/methods , Hippocampus/physiology , Hippocampus/radiation effects , Receptors, Nicotinic/physiology , Aconitine/pharmacology , Animals , Animals, Newborn , Bicuculline/pharmacology , Biological Clocks/drug effects , Biological Clocks/physiology , Bungarotoxins/pharmacology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electrophysiology/methods , GABA Antagonists/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , In Vitro Techniques , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Rats , Rats, Wistar , Spectrum Analysis , Time Factors , alpha7 Nicotinic Acetylcholine Receptor
14.
Diabetes ; 51 Suppl 1: S25-8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11815453

ABSTRACT

Various fluorescent probes were assessed for investigating intact islets of Langerhans using two-photon excitation imaging. Polar fluorescent tracers applied on the outside rapidly (within 3 min) penetrated deep into the islets via microvessels. Likewise, an adenovirus carrying a Ca(2+)-sensitive green fluorescent protein mutant gene, yellow cameleon 2.1, was successfully transfected and enabled ratiometric cytosolic Ca(2+) measurement of cells in the deep layers of the islets. Interestingly, FM1-43, which is lipophilic and does not permeate the plasma membrane, also rapidly reached deep cell layers of the islets. In contrast, lipophilic fluorescent probes that permeate the plasma membrane (for example, fura-2-acetoxymethyl and BODIPY-forskolin) accumulated in the superficial cell layers of the islets, even 30 min after application. Thus, two-photon excitation imaging of pancreatic islets is a promising method for clarifying signaling mechanisms of islet cells, particularly when it is combined with membrane-impermeable probes. In addition, our data suggest that membrane-permeable antagonists may affect only the superficial cell layers of islets, and so their negative effects should be interpreted with caution.


Subject(s)
Fluorescent Dyes/pharmacokinetics , Islets of Langerhans/physiology , Microscopy, Fluorescence/methods , Animals , Boron Compounds/pharmacokinetics , Colforsin/pharmacokinetics , Fura-2/analogs & derivatives , Fura-2/pharmacokinetics , Isoquinolines/pharmacokinetics , Lipids , Mice , Photons , Pyridinium Compounds/pharmacokinetics , Quaternary Ammonium Compounds/pharmacokinetics , Water
SELECTION OF CITATIONS
SEARCH DETAIL
...