Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Sci Rep ; 11(1): 5552, 2021 03 10.
Article En | MEDLINE | ID: mdl-33692389

Retinoid X receptors are members of the nuclear receptor family that regulate gene expression in response to retinoic acid and related ligands. Group 1 metabotropic glutamate receptors are G-protein coupled transmembrane receptors that activate intracellular signaling cascades in response to the neurotransmitter, glutamate. These two classes of molecules have been studied independently and found to play important roles in regulating neuronal physiology with potential clinical implications for disorders such as depression, schizophrenia, Parkinson's and Alzheimer's disease. Here we show that mice lacking the retinoid X receptor subunit, RXRγ, exhibit impairments in group 1 mGluR-mediated electrophysiological responses at hippocampal Schaffer collateral-CA1 pyramidal cell synapses, including impaired group 1 mGluR-dependent long-term synaptic depression (LTD), reduced group 1 mGluR-induced calcium release, and loss of group 1 mGluR-activated voltage-sensitive currents. These animals also exhibit impairments in a subset of group 1 mGluR-dependent behaviors, including motor performance, spatial object recognition, and prepulse inhibition. Together, these observations demonstrate convergence between the RXRγ and group 1 mGluR signaling pathways that may function to coordinate their regulation of neuronal activity. They also identify RXRγ as a potential target for the treatment of disorders in which group 1 mGluR signaling has been implicated.


CA1 Region, Hippocampal/metabolism , Long-Term Synaptic Depression , Pyramidal Cells/metabolism , Receptors, Metabotropic Glutamate/metabolism , Retinoid X Receptor gamma/metabolism , Signal Transduction , Synapses/metabolism , Animals , Mice , Mice, Knockout , Receptors, Metabotropic Glutamate/genetics , Retinoid X Receptor gamma/genetics , Synapses/genetics
2.
J Alzheimers Dis ; 79(4): 1813-1829, 2021.
Article En | MEDLINE | ID: mdl-33459709

BACKGROUND: The serine/threonine protein phosphatase, PP2A, is thought to play a central role in the molecular pathogenesis of Alzheimer's disease (AD), and the activity and substrate specificity of PP2A is regulated, in part, through methylation and demethylation of its catalytic subunit. Previously, we found that transgenic overexpression of the PP2A methyltransferase, LCMT-1, or the PP2A methylesterase, PME-1, altered the sensitivity of mice to impairments caused by acute exposure to synthetic oligomeric amyloid-ß (Aß). OBJECTIVE: Here we sought to test the possibility that these molecules also controlled sensitivity to impairments caused by chronically elevated levels of Aß produced in vivo. METHODS: To do this, we examined the effects of transgenic LCMT-1, or PME-1 overexpression on cognitive and electrophysiological impairments caused by chronic overexpression of mutant human APP in Tg2576 mice. RESULTS: We found that LCMT-1 overexpression prevented impairments in short-term spatial memory and synaptic plasticity in Tg2576 mice, without altering APP expression or soluble Aß levels. While the magnitude of the effects of PME-1 overexpression in Tg2576 mice was small and potentially confounded by the emergence of non-cognitive impairments, Tg2576 mice that overexpressed PME-1 showed a trend toward earlier onset and/or increased severity of cognitive and electrophysiological impairments. CONCLUSION: These data suggest that the PP2A methyltransferase, LCMT-1, and the PP2A methylesterase, PME-1, may participate in the molecular pathogenesis of AD by regulating sensitivity to the pathogenic effects of chronically elevated levels of Aß.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Carboxylic Ester Hydrolases/metabolism , Protein O-Methyltransferase/metabolism , Alzheimer Disease/complications , Amyloid beta-Peptides/genetics , Animals , Cognitive Dysfunction/etiology , Disease Models, Animal , Humans , Mice , Mice, Transgenic
3.
J Neurosci ; 40(23): 4596-4608, 2020 06 03.
Article En | MEDLINE | ID: mdl-32341098

Beta-amyloid (Aß) is thought to play a critical role in Alzheimer's disease (AD), and application of soluble oligomeric forms of Aß produces AD-like impairments in cognition and synaptic plasticity in experimental systems. We found previously that transgenic overexpression of the PP2A methylesterase, PME-1, or the PP2A methyltransferase, LCMT-1, altered the sensitivity of mice to Aß-induced impairments, suggesting that PME-1 inhibition may be an effective approach for preventing or treating these impairments. To explore this possibility, we examined the behavioral and electrophysiological effects of acutely applied synthetic Aß oligomers in male and female mice heterozygous for either a PME-1 KO or an LCMT-1 gene-trap mutation. We found that heterozygous PME-1 KO mice were resistant to Aß-induced impairments in cognition and synaptic plasticity, whereas LCMT-1 gene-trap mice showed increased sensitivity to Aß-induced impairments. The heterozygous PME-1 KO mice produced normal levels of endogenous Aß and exhibited normal electrophysiological responses to picomolar concentrations of Aß, suggesting that reduced PME-1 expression in these animals protects against Aß-induced impairments without impacting normal physiological Aß functions. Together, these data provide additional support for roles for PME-1 and LCMT-1 in regulating sensitivity to Aß-induced impairments, and suggest that inhibition of PME-1 may constitute a viable therapeutic approach for selectively protecting against the pathologic actions of Aß in AD.SIGNIFICANCE STATEMENT Elevated levels of ß-amyloid (Aß) in the brain are thought to contribute to the cognitive impairments observed in Alzheimer's disease patients. Here we show that genetically reducing endogenous levels of the PP2A methylesterase, PME-1, prevents the cognitive and electrophysiological impairments caused by acute exposure to pathologic concentrations of Aß without impairing normal physiological Aß function or endogenous Aß production. Conversely, reducing endogenous levels of the PP2A methyltransferase, LCMT-1, increases sensitivity to Aß-induced impairments. These data offer additional insights into the molecular factors that control sensitivity to Aß-induced impairments, and suggest that inhibiting PME-1 may constitute a viable therapeutic avenue for preventing Aß-related impairments in Alzheimer's disease.


Amyloid beta-Peptides/toxicity , Carboxylic Ester Hydrolases/biosynthesis , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/enzymology , Protein O-Methyltransferase/biosynthesis , Animals , Carboxylic Ester Hydrolases/genetics , Cognitive Dysfunction/physiopathology , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Female , Gene Expression , Male , Mice , Mice, Knockout , Protein O-Methyltransferase/genetics
4.
Methods Mol Biol ; 1779: 85-97, 2018.
Article En | MEDLINE | ID: mdl-29886529

Oligomerization of soluble tau protein is attracting the attention of an increasingly larger number of scientists involved in research on Alzheimer's disease and other tauopathies. A variety of methods have been developed for the purification of proteins from biological tissues and bacterial cells. Various types of high performance liquid chromatography (HPLC) and affinity tags represent the most common techniques for isolating proteins. Here, we describe a procedure for extracting recombinant tau protein from bacterial cells, utilizing a 6×His affinity tag, or endogenous tau from brain cortices using acid extraction followed by fast protein liquid chromatography (FPLC). Additionally, we introduce a method for oligomerization based on reduction and oxidation of cysteine residues. Our preparation assures high yield of tau protein, while preserving its physiological function.


Bacteria/metabolism , Brain/metabolism , tau Proteins/chemistry , tau Proteins/isolation & purification , Alzheimer Disease/metabolism , Animals , Autopsy , Bacteria/genetics , Chromatography, Affinity , Chromatography, High Pressure Liquid , Cysteine/chemistry , Humans , Mice , Oxidation-Reduction , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , tau Proteins/genetics
5.
PLoS One ; 12(12): e0189413, 2017.
Article En | MEDLINE | ID: mdl-29253878

Soluble forms of oligomeric beta-amyloid (Aß) are thought to play a central role in Alzheimer's disease (AD). Transgenic manipulation of methylation of the serine/threonine protein phosphatase, PP2A, was recently shown to alter the sensitivity of mice to AD-related impairments resulting from acute exposure to elevated levels of Aß. In addition, eicosanoyl-5-hydroxytryptamide (EHT), a naturally occurring component from coffee beans that modulates PP2A methylation, was shown to confer therapeutic benefits in rodent models of AD and Parkinson's disease. Here, we tested the hypothesis that EHT protects animals from the pathological effects of exposure to elevated levels of soluble oligomeric Aß. We treated mice with EHT-containing food at two different doses and assessed the sensitivity of these animals to Aß-induced behavioral and electrophysiological impairments. We found that EHT administration protected animals from Aß-induced cognitive impairments in both a radial-arm water maze and contextual fear conditioning task. We also found that both chronic and acute EHT administration prevented Aß-induced impairments in long-term potentiation. These data add to the accumulating evidence suggesting that interventions with pharmacological agents, such as EHT, that target PP2A activity may be therapeutically beneficial for AD and other neurological conditions.


Alzheimer Disease/drug therapy , Amyloid beta-Peptides/chemistry , Cognition Disorders/prevention & control , Serotonin/analogs & derivatives , Alzheimer Disease/pathology , Animals , Coffee , Cognition/drug effects , Conditioning, Psychological , Disease Models, Animal , Electrophysiology , Fear , Female , Long-Term Potentiation , Male , Maze Learning , Methylation , Mice , Mice, Inbred C57BL , Nervous System Diseases/drug therapy , Nervous System Diseases/pathology , Neuronal Plasticity , Phosphorylation , Serotonin/pharmacology , Solubility
6.
Proc Natl Acad Sci U S A ; 113(12): 3347-52, 2016 Mar 22.
Article En | MEDLINE | ID: mdl-26951658

Elevated levels of the ß-amyloid peptide (Aß) are thought to contribute to cognitive and behavioral impairments observed in Alzheimer's disease (AD). Protein phosphatase 2A (PP2A) participates in multiple molecular pathways implicated in AD, and its expression and activity are reduced in postmortem brains of AD patients. PP2A is regulated by protein methylation, and impaired PP2A methylation is thought to contribute to increased AD risk in hyperhomocysteinemic individuals. To examine further the link between PP2A and AD, we generated transgenic mice that overexpress the PP2A methylesterase, protein phosphatase methylesterase-1 (PME-1), or the PP2A methyltransferase, leucine carboxyl methyltransferase-1 (LCMT-1), and examined the sensitivity of these animals to behavioral and electrophysiological impairments caused by exogenous Aß exposure. We found that PME-1 overexpression enhanced these impairments, whereas LCMT-1 overexpression protected against Aß-induced impairments. Neither transgene affected Aß production or the electrophysiological response to low concentrations of Aß, suggesting that these manipulations selectively affect the pathological response to elevated Aß levels. Together these data identify a molecular mechanism linking PP2A to the development of AD-related cognitive impairments that might be therapeutically exploited to target selectively the pathological effects caused by elevated Aß levels in AD patients.


Amyloid beta-Peptides/physiology , Cognition Disorders/physiopathology , Protein Phosphatase 2/metabolism , Animals , Behavior, Animal , Methylation , Mice , Mice, Transgenic
7.
J Neurotrauma ; 33(13): 1202-11, 2016 07 01.
Article En | MEDLINE | ID: mdl-26414212

An increasing number of studies have reported blood-brain barrier (BBB) dysfunction after blast-induced traumatic brain injury (bTBI). Despite this evidence, there is limited quantitative understanding of the extent of BBB opening and the time course of damage after blast injury. In addition, many studies do not report kinematic parameters of head motion, making it difficult to separate contributions of primary and tertiary blast-loading. Detailed characterization of blast-induced BBB damage may hold important implications for serum constituents that may potentially cross the compromised barrier and contribute to neurotoxicity, neuroinflammation, and persistent neurologic deficits. Using an in vivo bTBI model, systemic administration of sodium fluorescein (NaFl; 376 Da), Evans blue (EB; 69 kDa when bound to serum albumin), and dextrans (3-500 kDa) was used to estimate the pore size of BBB opening and the time required for recovery. Exposure to blast with 272 ± 6 kPa peak overpressure, 0.69 ± 0.01 ms duration, and 65 ± 1 kPa*ms impulse resulted in significant acute extravasation of NaFl, 3 kDa dextran, and EB. However, there was no significant acute extravasation of 70 kDa or 500 kDa dextrans, and minimal to no extravasation of NaFl, dextrans, or EB 1 day after exposure. This study presents a detailed analysis of the time course and pore size of BBB opening after bTBI, supported by a characterization of kinematic parameters associated with blast-induced head motion.


Blast Injuries/complications , Blood-Brain Barrier/injuries , Blood-Brain Barrier/physiopathology , Brain Injuries, Traumatic/complications , Disease Models, Animal , Animals , Female , Male , Mice , Mice, Inbred C57BL
8.
Neurotherapeutics ; 12(1): 29-41, 2015 Jan.
Article En | MEDLINE | ID: mdl-25575647

Therapeutic attempts to cure Alzheimer's disease (AD) have failed, and new strategies are desperately needed. Motivated by this reality, many laboratories (including our own) have focused on synaptic dysfunction in AD because synaptic changes are highly correlated with the severity of clinical dementia. In particular, memory formation is accompanied by altered synaptic strength, and this phenomenon (and its dysfunction in AD) has been a recent focus for many laboratories. The molecule cyclic adenosine monophosphate response element-binding protein (CREB) is at a central converging point of pathways and mechanisms activated during the processes of synaptic strengthening and memory formation, as CREB phosphorylation leads to transcription of memory-associated genes. Disruption of these mechanisms in AD results in a reduction of CREB activation with accompanying memory impairment. Thus, it is likely that strategies aimed at these mechanisms will lead to future therapies for AD. In this review, we will summarize literature that investigates 5 possible therapeutic pathways for rescuing synaptic dysfunction in AD: 4 enzymatic pathways that lead to CREB phosphorylation (the cyclic adenosine monophosphate cascade, the serine/threonine kinases extracellular regulated kinases 1 and 2, the nitric oxide cascade, and the calpains), as well as histone acetyltransferases and histone deacetylases (2 enzymes that regulate the histone acetylation necessary for gene transcription).


Alzheimer Disease/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Synaptic Transmission/physiology , Alzheimer Disease/pathology , Animals , Humans , Synapses/metabolism , Synapses/pathology
9.
Neuron ; 58(1): 104-17, 2008 Apr 10.
Article En | MEDLINE | ID: mdl-18400167

While most studies have focused on the role of long-term potentiation in behavior, far less is known about the role of long-term depression (LTD). To examine the potential involvement of LTD in learning and memory, we generated transgenic mice that express a fragment of the SV40 small t antigen known to inhibit protein phosphatase 2A (PP2A). Small t antigen expression blocked both stimulus-induced and chemically induced NMDAR-dependent LTD at Schaffer collateral synapses but did not affect potentiation, depotentiation, or mGluR-dependent LTD. This physiological phenotype was associated with deficits in behavioral flexibility in both the Morris water maze and a delayed nonmatch to place T-maze task, suggesting that NMDAR-dependent LTD is required for behavioral flexibility and may act by weakening previously encoded memory traces when new information is learned.


Long-Term Synaptic Depression/genetics , Maze Learning/physiology , Receptors, N-Methyl-D-Aspartate/deficiency , Receptors, N-Methyl-D-Aspartate/genetics , Adaptation, Psychological/physiology , Animals , Behavior, Animal/physiology , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Receptors, N-Methyl-D-Aspartate/physiology
10.
Learn Mem ; 15(4): 261-70, 2008 Apr.
Article En | MEDLINE | ID: mdl-18391187

Cyclic AMP signaling plays a central role in regulating activity at a number of synapses in the brain. We showed previously that pairing activation of receptors that inhibit adenylate cyclase (AC) and reduce the concentration of cyclic AMP, with elevation of the concentration of cyclic GMP is sufficient to elicit a presynaptically expressed form of LTD at Schaffer collateral-CA1 synapses in the hippocampus. To directly test the role of AC inhibition and G-protein signaling in LTD at these synapses, we utilized transgenic mice that express a mutant, constitutively active inhibitory G protein, Galpha(i2), in principal neurons of the forebrain. Transgene expression of Galpha(i2) markedly enhanced LTD and impaired late-phase LTP at Schaffer collateral synapses, with no associated differences in input/output relations, paired-pulse facilitation, or NMDA receptor-gated conductances. When paired with application of a type V phosphodiesterase inhibitor to elevate the concentration of intracellular cyclic GMP, constitutively active Galpha(i2) expression converted the transient depression normally caused by this treatment to an LTD that persisted after the drug was washed out. Moreover, this effect could be mimicked in control slices by pairing type V phosphodiesterase inhibitor application with application of a PKA inhibitor. Electrophysiological recordings of spontaneous excitatory postsynaptic currents and two-photon visualization of vesicular release using FM1-43 revealed that constitutively active Galpha(i2) tonically reduced basal release probability from the rapidly recycling vesicle pool of Schaffer collateral terminals. Our findings support the hypothesis that inhibitory G-protein signaling acts presynaptically to regulate release, and, when paired with elevations in the concentration of cyclic GMP, converts a transient cyclic GMP-induced depression into a long-lasting decrease in release.


Adenylyl Cyclase Inhibitors , Cyclic GMP/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Protein Regulators/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , Hippocampus , Intracellular Signaling Peptides and Proteins/pharmacology , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Receptors, Presynaptic/drug effects , Synapses/drug effects , Animals , Depression/metabolism , Depression/psychology , Disease Models, Animal , Drug Synergism , Enzyme Inhibitors/administration & dosage , Hippocampus/anatomy & histology , Hippocampus/drug effects , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/administration & dosage , Long-Term Potentiation , Mice , Mice, Transgenic , Phosphodiesterase Inhibitors/administration & dosage
11.
Proc Natl Acad Sci U S A ; 103(16): 6380-5, 2006 Apr 18.
Article En | MEDLINE | ID: mdl-16606834

Presynaptic inhibitory G protein-coupled receptors play a critical role in regulating transmission at a number of synapses in the central and peripheral nervous system. We generated transgenic mice that express a constitutively active form of an inhibitory Galpha subunit to examine the molecular mechanisms underlying the actions of one such receptor, metabotropic glutamate receptor (mGluR) 2, at mossy fiber-CA3 synapses in the hippocampus. mGluR2 participates in at least three types of mossy fiber synaptic plasticity, (i) transient suppression of synaptic transmission, (ii) long-term depression (LTD), and (iii) inhibition of long-term potentiation (LTP), and we find that inhibitory Galpha signaling is sufficient to account for the actions of mGluR2 in each. The fact that constitutively active Galphai2 occludes the transient suppression of synaptic transmission by mGluR2, while enhancing LTD, suggests further that these two forms of plasticity are expressed via different mechanisms. In addition, the LTP deficit observed in constitutively active Galphai2-expressing mice suggests that mGluR2 activation may serve as a metaplastic switch to permit the induction of LTD by inhibiting LTP.


GTP-Binding Protein alpha Subunit, Gi2/metabolism , Long-Term Synaptic Depression , Mossy Fibers, Hippocampal/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synapses/metabolism , Synaptic Transmission , Adenylyl Cyclase Inhibitors , Animals , GTP-Binding Protein alpha Subunit, Gi2/genetics , Hippocampus/chemistry , Hippocampus/metabolism , Long-Term Synaptic Depression/genetics , Mice , Mice, Transgenic , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Synaptic Transmission/genetics
...