Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Comp Neurol ; 527(4): 856-873, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30408169

ABSTRACT

Female rhesus monkeys and women are subject to age- and menopause-related deficits in working memory, an executive function mediated by the dorsolateral prefrontal cortex (dlPFC). Long-term cyclic administration of 17ß-estradiol improves working memory, and restores highly plastic axospinous synapses within layer III dlPFC of aged ovariectomized monkeys. In this study, we tested the hypothesis that synaptic distributions of tau protein phosphorylated at serine 214 (pS214-tau) are altered with age or estradiol treatment, and couple to working memory performance. First, ovariectormized young and aged monkeys received vehicle or estradiol treatment, and were tested on the delayed response (DR) test of working memory. Serial section electron microscopic immunocytochemistry was then performed to quantitatively assess the subcellular synaptic distributions of pS214-tau. Overall, the majority of synapses contained pS214-tau immunogold particles, which were predominantly localized to the cytoplasm of axon terminals. pS214-tau was also abundant within synaptic and cytoplasmic domains of dendritic spines. The density of pS214-tau immunogold within the active zone, cytoplasmic, and plasmalemmal domains of axon terminals, and subjacent to the postsynaptic density within the subsynaptic domains of dendritic spines, were each reduced with age. None of the variables examined were directly linked to cognitive status, but a high density of pS214-tau immunogold particles within presynaptic cytoplasmic and plasmalemmal domains, and within postsynaptic subsynaptic and plasmalemmal domains, accompanied high synapse density. Together, these data support a possible physiological, rather than pathological, role for pS214-tau in the modulation of synaptic morphology in monkey dlPFC.


Subject(s)
Aging/metabolism , Dendritic Spines/metabolism , Prefrontal Cortex/metabolism , Synapses/metabolism , tau Proteins/metabolism , Aging/pathology , Animals , Cognitive Dysfunction/metabolism , Dendritic Spines/pathology , Female , Macaca mulatta , Memory, Short-Term/physiology , Synapses/pathology
2.
Neuroscience ; 394: 303-315, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30482274

ABSTRACT

Age- and menopause-related deficits in working memory can be partially restored with estradiol replacement in women and female nonhuman primates. Working memory is a cognitive function reliant on persistent firing of dorsolateral prefrontal cortex (dlPFC) neurons that requires the activation of GluN2B-containing glutamate NMDA receptors. We tested the hypothesis that the distribution of phospho-Tyr1472-GluN2B (pGluN2B), a predominant form of GluN2B seen at the synapse, is sensitive to aging or estradiol treatment and coupled to working memory performance. First, ovariectomized young and aged rhesus monkeys (Macaca mulatta) received long-term cyclic vehicle (V) or estradiol (E) treatment and were tested on the delayed response (DR) test of working memory. Then, serial section electron microscopic immunocytochemistry was performed to quantitatively assess the subcellular distribution of pGluN2B. While the densities of pGluN2B immunogold particles in dlPFC dendritic spines were not different across age or treatment groups, the percentage of gold particles located within the synaptic compartment was significantly lower in aged-E monkeys compared to young-E and aged-V monkeys. On the other hand, the percentage of pGluN2B gold particles in the spine cytoplasm was decreased with E treatment in young, but increased with E in aged monkeys. In aged monkeys, DR average accuracy inversely correlated with the percentage of synaptic pGluN2B, while it positively correlated with the percentage of cytoplasmic pGluN2B. Together, E replacement may promote cognitive health in aged monkeys, in part, by decreasing the relative representation of synaptic pGluN2B and potentially protecting the dlPFC from calcium toxicity.


Subject(s)
Aging , Estrogens/administration & dosage , Memory, Short-Term/physiology , Prefrontal Cortex/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology , Animals , Dendritic Spines/drug effects , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Female , Macaca mulatta , Memory, Short-Term/drug effects , Phosphorylation , Post-Synaptic Density/ultrastructure , Prefrontal Cortex/drug effects , Prefrontal Cortex/ultrastructure , Receptors, N-Methyl-D-Aspartate/ultrastructure , Synapses/drug effects , Synapses/ultrastructure
3.
Neuron ; 98(6): 1064-1066, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29953868

ABSTRACT

Drug treatments to improve memory focus on enhancing acetylcholine. However, Vijayraghavan and colleagues (2018) show that direct stimulation of the M1 muscarinic acetylcholine receptor adversely affected neuronal activity in prefrontal cortex related to working memory for behavioral rules.


Subject(s)
Memory, Short-Term , Receptor, Muscarinic M1 , Animals , Cognition , Prefrontal Cortex , Primates
4.
Cereb Cortex ; 28(4): 1458-1471, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351585

ABSTRACT

Neurons in primary visual cortex (V1) are more resilient than those in dorsolateral prefrontal cortex (dlPFC) in aging, schizophrenia and Alzheimer's disease. The current study compared glutamate and neuromodulatory actions in macaque V1 to those in dlPFC, and found striking regional differences. V1 neuronal firing to visual stimuli depended on AMPA receptors, with subtle NMDA receptor contributions, while dlPFC depends primarily on NMDA receptors. Neuromodulatory actions also differed between regions. In V1, cAMP signaling increased neuronal firing, and the phosphodiesterase PDE4A was positioned to regulate cAMP effects on glutamate release from axons. HCN channels in V1 were classically located on distal dendrites, and enhanced cell firing. These data contrast with dlPFC, where PDE4A and HCN channels are concentrated in thin spines, and cAMP-HCN signaling gates inputs and weakens firing. These regional differences may explain why V1 neurons are more resilient than dlPFC neurons to the challenges of age and disease.


Subject(s)
Nerve Net/physiology , Neurons/physiology , Prefrontal Cortex/cytology , Synapses/physiology , Visual Cortex/cytology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cardiovascular Agents/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/ultrastructure , Dendritic Spines/drug effects , Dendritic Spines/ultrastructure , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/ultrastructure , Macaca mulatta , Membrane Potentials/drug effects , Microscopy, Immunoelectron , Nerve Net/drug effects , Neurons/drug effects , Neurons/ultrastructure , Photic Stimulation , Pyrimidines/pharmacology , Signal Transduction/drug effects , Synapses/drug effects , Synapses/ultrastructure
5.
Alzheimers Dement ; 14(5): 680-691, 2018 05.
Article in English | MEDLINE | ID: mdl-29241829

ABSTRACT

INTRODUCTION: An animal model of late-onset Alzheimer's disease is needed to research what causes degeneration in the absence of dominant genetic insults and why the association cortex is particularly vulnerable to degeneration. METHODS: We studied the progression of tau and amyloid cortical pathology in the aging rhesus macaque using immunoelectron microscopy and biochemical assays. RESULTS: Aging macaques exhibited the same qualitative pattern and sequence of tau and amyloid cortical pathology as humans, reaching Braak stage III/IV. Pathology began in the young-adult entorhinal cortex with protein kinase A-phosphorylation of tau, progressing to fibrillation with paired helical filaments and mature tangles in oldest animals. Tau pathology in the dorsolateral prefrontal cortex paralleled but lagged behind the entorhinal cortex, not afflicting the primary visual cortex. DISCUSSION: The aging rhesus macaque provides the long-sought animal model for exploring the etiology of late-onset Alzheimer's disease and for testing preventive strategies.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Disease Models, Animal , Disease Progression , Macaca mulatta , Amyloid/metabolism , Animals , Brain/pathology , Entorhinal Cortex/pathology , Microscopy, Immunoelectron/methods , Neurofibrillary Tangles/pathology , Phosphorylation , Plaque, Amyloid/pathology , Prefrontal Cortex , tau Proteins/metabolism
6.
Cereb Cortex ; 27(3): 2022-2033, 2017 03 01.
Article in English | MEDLINE | ID: mdl-26941383

ABSTRACT

Age- and menopause-related impairment in working memory mediated by the dorsolateral prefrontal cortex (dlPFC) occurs in humans and nonhuman primates. Long-term cyclic 17ß-estradiol treatment rescues cognitive deficits in aged ovariectomized rhesus monkeys while restoring highly plastic synapses. Here we tested whether distributions of G protein-coupled estrogen receptor 1 (GPER1) within monkey layer III dlPFC synapses are sensitive to age and estradiol, and coupled to cognitive function. Ovariectomized young and aged monkeys administered vehicle or estradiol were first tested on a delayed response test of working memory. Then, quantitative serial section immunoelectron microscopy was used to determine the distributions of synaptic GPER1. GPER1-containing nonperforated axospinous synapse density was reduced with age, and partially restored with estrogen treatment. The majority of synapses expressed GPER1, which was predominately localized to presynaptic cytoplasm and mitochondria. GPER1 was also abundant at plasmalemmas, and within cytoplasmic and postsynaptic density (PSD) domains of dendritic spines. GPER1 levels did not differ with age or treatment, and none of the variables examined were tightly associated with cognitive function. However, greater representation of GPER1 subjacent to the PSD accompanied higher synapse density. These data suggest that GPER1 is positioned to support diverse functions key to synaptic plasticity in monkey dlPFC.


Subject(s)
Aging/metabolism , Estrogen Receptor alpha/metabolism , Menopause/metabolism , Prefrontal Cortex/metabolism , Synapses/metabolism , Aging/pathology , Animals , Estradiol/administration & dosage , Estrogens/administration & dosage , Female , Hormone Replacement Therapy , Immunohistochemistry , Macaca mulatta , Microscopy, Immunoelectron , Neuronal Plasticity/physiology , Ovariectomy , Prefrontal Cortex/pathology , Synapses/pathology
7.
Cereb Cortex ; 25(6): 1454-68, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24323499

ABSTRACT

The effects of normal aging on morphologic and electrophysiologic properties of layer 3 pyramidal neurons in rhesus monkey primary visual cortex (V1) were assessed with whole-cell, patch-clamp recordings in in vitro slices. In another cohort of monkeys, the ultrastructure of synapses in the layers 2-3 neuropil of V1 was assessed using electron microscopy. Distal apical dendritic branching complexity was reduced in aged neurons, as was the total spine density, due to specific loss of mushroom spines from the apical tree and of thin spines from the basal tree. There was also an age-related decrease in the numerical density of symmetric and asymmetric synapses. In contrast to these structural changes, intrinsic membrane, action potential (AP), and excitatory and inhibitory synaptic current properties were the same in aged and young neurons. Computational modeling using morphologic reconstructions predicts that reduced dendritic complexity leads to lower attenuation of voltage outward from the soma (e.g., backpropagating APs) in aged neurons. Importantly, none of the variables that changed with age differed in neurons from cognitively impaired versus unimpaired aged monkeys. In summary, there are age-related alterations to the structural properties of V1 neurons, but these are not associated with significant electrophysiologic changes or with cognitive decline.


Subject(s)
Aging , Cognition/physiology , Pyramidal Cells/physiology , Visual Cortex/cytology , Animals , Computer Simulation , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Electric Stimulation , Excitatory Postsynaptic Potentials/physiology , Female , Lysine/analogs & derivatives , Macaca mulatta , Male , Membrane Potentials/physiology , Microscopy, Electron , Models, Neurological , Neuropsychological Tests , Patch-Clamp Techniques , Pyramidal Cells/ultrastructure , Synapses/ultrastructure
8.
Ageing Res Rev ; 12(3): 757-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23528367

ABSTRACT

The synaptic connections that form between neurons during development remain plastic and able to adapt throughout the lifespan, enabling learning and memory. However, during aging and in particular in neurodegenerative diseases, synapses become dysfunctional and degenerate, contributing to dementia. In the case of Alzheimer's disease (AD), synapse loss is the strongest pathological correlate of cognitive decline, indicating that synaptic degeneration plays a central role in dementia. Over the past decade, strong evidence has emerged that oligomeric forms of amyloid beta, the protein that accumulates in senile plaques in the AD brain, contribute to degeneration of synaptic structure and function. More recent data indicate that pathological forms of tau protein, which accumulate in neurofibrillary tangles in the AD brain, also cause synaptic dysfunction and loss. In this review, we will present the case that soluble forms of both amyloid beta and tau protein act at the synapse to cause neural network dysfunction, and further that these two pathological proteins may act in concert to cause synaptic pathology. These data may have wide-ranging implications for the targeting of soluble pathological proteins in neurodegenerative diseases to prevent or reverse cognitive decline.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Synapses/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Humans
9.
J Neurosci ; 32(40): 13644-60, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23035077

ABSTRACT

Whole-cell patch-clamp recordings and high-resolution 3D morphometric analyses of layer 3 pyramidal neurons in in vitro slices of monkey primary visual cortex (V1) and dorsolateral granular prefrontal cortex (dlPFC) revealed that neurons in these two brain areas possess highly distinctive structural and functional properties. Area V1 pyramidal neurons are much smaller than dlPFC neurons, with significantly less extensive dendritic arbors and far fewer dendritic spines. Relative to dlPFC neurons, V1 neurons have a significantly higher input resistance, depolarized resting membrane potential, and higher action potential (AP) firing rates. Most V1 neurons exhibit both phasic and regular-spiking tonic AP firing patterns, while dlPFC neurons exhibit only tonic firing. Spontaneous postsynaptic currents are lower in amplitude and have faster kinetics in V1 than in dlPFC neurons, but are no different in frequency. Three-dimensional reconstructions of V1 and dlPFC neurons were incorporated into computational models containing Hodgkin-Huxley and AMPA receptor and GABA(A) receptor gated channels. Morphology alone largely accounted for observed passive physiological properties, but led to AP firing rates that differed more than observed empirically, and to synaptic responses that opposed empirical results. Accordingly, modeling predicts that active channel conductances differ between V1 and dlPFC neurons. The unique features of V1 and dlPFC neurons are likely fundamental determinants of area-specific network behavior. The compact electrotonic arbor and increased excitability of V1 neurons support the rapid signal integration required for early processing of visual information. The greater connectivity and dendritic complexity of dlPFC neurons likely support higher level cognitive functions including working memory and planning.


Subject(s)
Neurons/physiology , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Visual Cortex/physiology , Action Potentials , Animals , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials/physiology , Female , In Vitro Techniques , Inhibitory Postsynaptic Potentials/physiology , Macaca mulatta , Male , Microscopy, Confocal , Models, Neurological , Neurons/ultrastructure , Organ Specificity , Patch-Clamp Techniques , Prefrontal Cortex/cytology , Synaptic Transmission , Visual Cortex/cytology
10.
Acta Neuropathol ; 124(6): 777-95, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22976049

ABSTRACT

Whole-cell patch-clamp recordings and high-resolution morphometry were used to assess functional and structural properties of layer 3 pyramidal neurons in early (<4 months) and advanced (>8 months) stages of tauopathy in frontal cortical slices prepared from rTg4510 tau mutant (P301L) mice. In early tauopathy, dendritic architecture is preserved. In advanced tauopathy, neurons can be categorized as either "atrophic" (58 %)-exhibiting marked atrophy of the apical tuft, or "intact" (42 %)-with normal apical tufts and, in some instances, proliferative sprouting of oblique branches of the apical trunk. Approximately equal numbers of atrophic and intact neurons contain neurofibrillary tangles (NFTs) or are tangle-free, lending further support to the idea that NFTs per se are not toxic. Spine density is decreased due to a specific reduction in mushroom spines, but filopodia are increased in both atrophic and intact neurons. By contrast to these morphological changes, which are robust only in the advanced stage, significant electrophysiological changes are present in the early stage and persist in the advanced stage in both atrophic and intact neurons. The most marked of these changes are: a depolarized resting membrane potential, an increased depolarizing sag potential and increased action potential firing rates-all indicative of hyperexcitability. Spontaneous excitatory postsynaptic currents are not reduced in frequency or amplitude in either stage. The difference in the time course of functionally important electrophysiological changes versus regressive morphological changes implies differences in pathogenic mechanisms underlying functional and structural changes to neurons during progressive tauopathy.


Subject(s)
Electrophysiological Phenomena/physiology , Frontal Lobe/pathology , Membrane Potentials/physiology , Pyramidal Cells/pathology , Tauopathies/physiopathology , Action Potentials/physiology , Animals , Dendrites/pathology , Dendrites/physiology , Disease Models, Animal , Excitatory Postsynaptic Potentials/physiology , Mice , Mice, 129 Strain , Mice, Transgenic , Patch-Clamp Techniques/methods , Pyramidal Cells/physiology , Tauopathies/pathology
11.
Acta Neuropathol ; 122(5): 551-64, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21968531

ABSTRACT

Cortical neuron death is prevalent by 9 months in rTg(tau(P301L))4510 tau mutant mice (TG) and surviving pyramidal cells exhibit dendritic regression and spine loss. We used whole-cell patch-clamp recordings to investigate the impact of these marked structural changes on spontaneous excitatory and inhibitory postsynaptic currents (sEPSCs and sIPSCs) of layer 3 pyramidal cells in frontal cortical slices from behaviorally characterized TG and non-transgenic (NT) mice at this age. Frontal lobe function of TG mice was intact following a short delay interval but impaired following a long delay interval in an object recognition test, and cortical atrophy and cell loss were pronounced. Surviving TG cells had significantly reduced dendritic diameters, total spine density, and mushroom spines, yet sEPSCs were increased and sIPSCs were unchanged in frequency. Thus, despite significant regressive structural changes, synaptic responses were not reduced in TG cells, indicating that homeostatic compensatory mechanisms occur during progressive tauopathy. Consistent with this idea, surviving TG cells were more intrinsically excitable than NT cells, and exhibited sprouting of filopodia and axonal boutons. Moreover, the neuropil in TG mice showed an increased density of asymmetric synapses, although their mean size was reduced. Taken together, these data indicate that during progressive tauopathy, cortical pyramidal cells compensate for loss of afferent input by increased excitability and establishment of new synapses. These compensatory homeostatic mechanisms may play an important role in slowing the progression of neuronal network dysfunction during neurodegenerative tauopathies.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Homeostasis/physiology , Inhibitory Postsynaptic Potentials/physiology , Pyramidal Cells/physiopathology , Tauopathies/physiopathology , Animals , Cognition/physiology , Disease Models, Animal , Disease Progression , Frontal Lobe/metabolism , Frontal Lobe/pathology , Mice , Mice, Mutant Strains , Patch-Clamp Techniques , Pyramidal Cells/pathology , Synapses/physiology , Tauopathies/pathology , tau Proteins/genetics , tau Proteins/metabolism
12.
Brain Struct Funct ; 214(2-3): 181-99, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20177698

ABSTRACT

In neurodegenerative disorders, such as Alzheimer's disease, neuronal dendrites and dendritic spines undergo significant pathological changes. Because of the determinant role of these highly dynamic structures in signaling by individual neurons and ultimately in the functionality of neuronal networks that mediate cognitive functions, a detailed understanding of these changes is of paramount importance. Mutant murine models, such as the Tg2576 APP mutant mouse and the rTg4510 tau mutant mouse have been developed to provide insight into pathogenesis involving the abnormal production and aggregation of amyloid and tau proteins, because of the key role that these proteins play in neurodegenerative disease. This review showcases the multidimensional approach taken by our collaborative group to increase understanding of pathological mechanisms in neurodegenerative disease using these mouse models. This approach includes analyses of empirical 3D morphological and electrophysiological data acquired from frontal cortical pyramidal neurons using confocal laser scanning microscopy and whole-cell patch-clamp recording techniques, combined with computational modeling methodologies. These collaborative studies are designed to shed insight on the repercussions of dystrophic changes in neocortical neurons, define the cellular phenotype of differential neuronal vulnerability in relevant models of neurodegenerative disease, and provide a basis upon which to develop meaningful therapeutic strategies aimed at preventing, reversing, or compensating for neurodegenerative changes in dementia.


Subject(s)
Alzheimer Disease/pathology , Cerebral Cortex/pathology , Dendrites/pathology , Electrophysiology/methods , Image Processing, Computer-Assisted/methods , Pyramidal Cells/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Computer Simulation , Dendrites/metabolism , Disease Models, Animal , Mice , Microscopy, Confocal/methods , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Patch-Clamp Techniques/methods , Pyramidal Cells/metabolism , Pyramidal Cells/physiopathology , Staining and Labeling/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...