Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 254: 109987, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38705570

ABSTRACT

Age is the largest risk factor for developing Alzheimer's disease (AD), a neurodegenerative disorder that causes a progressive and severe dementia. The underlying cause of cognitive deficits seen in AD is thought to be the disconnection of neural circuits that control memory and executive functions. Insight into the mechanisms by which AD diverges from normal aging will require identifying precisely which cellular events are driven by aging and which are impacted by AD-related pathologies. Since microglia, the brain-resident macrophages, are known to have critical roles in the formation and maintenance of neural circuits through synaptic pruning, they are well-positioned to modulate synaptic connectivity in circuits sensitive to aging or AD. In this review, we provide an overview of the current state of the field and on emerging technologies being employed to elucidate microglia-synaptic interactions in aging and AD. We also discuss the importance of leveraging genetic diversity to study how these interactions are shaped across more realistic contexts. We propose that these approaches will be essential to define specific aging- and disease-relevant trajectories for more personalized therapeutics aimed at reducing the effects of age or AD pathologies on the brain. This article is part of the Special Issue on "Microglia".


Subject(s)
Aging , Alzheimer Disease , Microglia , Synapses , Animals , Humans , Aging/physiology , Aging/pathology , Alzheimer Disease/pathology , Brain/pathology , Microglia/pathology , Microglia/physiology , Neuronal Plasticity/physiology , Synapses/pathology , Synapses/physiology
2.
Nat Commun ; 15(1): 2142, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38459070

ABSTRACT

Neuronal mitochondria play important roles beyond ATP generation, including Ca2+ uptake, and therefore have instructive roles in synaptic function and neuronal response properties. Mitochondrial morphology differs significantly between the axon and dendrites of a given neuronal subtype, but in CA1 pyramidal neurons (PNs) of the hippocampus, mitochondria within the dendritic arbor also display a remarkable degree of subcellular, layer-specific compartmentalization. In the dendrites of these neurons, mitochondria morphology ranges from highly fused and elongated in the apical tuft, to more fragmented in the apical oblique and basal dendritic compartments, and thus occupy a smaller fraction of dendritic volume than in the apical tuft. However, the molecular mechanisms underlying this striking degree of subcellular compartmentalization of mitochondria morphology are unknown, precluding the assessment of its impact on neuronal function. Here, we demonstrate that this compartment-specific morphology of dendritic mitochondria requires activity-dependent, Ca2+ and Camkk2-dependent activation of AMPK and its ability to phosphorylate two direct effectors: the pro-fission Drp1 receptor Mff and the recently identified anti-fusion, Opa1-inhibiting protein, Mtfr1l. Our study uncovers a signaling pathway underlying the subcellular compartmentalization of mitochondrial morphology in dendrites of neurons in vivo through spatially precise and activity-dependent regulation of mitochondria fission/fusion balance.


Subject(s)
Neurons , Pyramidal Cells , Neurons/metabolism , Pyramidal Cells/physiology , Hippocampus , Axons/metabolism , Mitochondria/metabolism , Dendrites/physiology
3.
Alzheimers Dement ; 20(1): 601-614, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37753835

ABSTRACT

INTRODUCTION: Human data suggest susceptibility and resilience to features of Alzheimer's disease (AD) such as microglia activation and synaptic dysfunction are under genetic control. However, causal relationships between these processes, and how genomic diversity modulates them remain systemically underexplored in mouse models. METHODS: AD-vulnerable hippocampal neurons were virally labeled in inbred (C57BL/6J) and wild-derived (PWK/PhJ) APP/PS1 and wild-type mice, and brain microglia depleted from 4 to 8 months of age. Dendrites were assessed for synapse plasticity changes by evaluating spine densities and morphologies. RESULTS: In C57BL/6J, microglia depletion blocked amyloid-induced synaptic density and morphology changes. At a finer scale, synaptic morphology on individual branches was dependent on microglia-dendrite physical interactions. Conversely, synapses from PWK/PhJ mice showed remarkable stability in response to amyloid, and no evidence of microglia contact-dependent changes on dendrites. DISCUSSION: These results demonstrate that microglia-dependent synaptic alterations in specific AD-vulnerable projection pathways are differentially controlled by genetic context.


Subject(s)
Alzheimer Disease , Humans , Mice , Animals , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Microglia/metabolism , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic , Mice, Inbred C57BL , Hippocampus/metabolism , Disease Models, Animal , Neuronal Plasticity/genetics , Synapses/metabolism , Amyloid/metabolism , Dendrites/metabolism
4.
Aging Cell ; 23(2): e14033, 2024 02.
Article in English | MEDLINE | ID: mdl-38130024

ABSTRACT

The disconnection of neuronal circuitry through synaptic loss is presumed to be a major driver of age-related cognitive decline. Age-related cognitive decline is heterogeneous, yet whether genetic mechanisms differentiate successful from unsuccessful cognitive decline through maintenance or vulnerability of synaptic connections remains unknown. Previous work using rodent and primate models leveraged various techniques to imply that age-related synaptic loss is widespread on pyramidal cells in prefrontal cortex (PFC) circuits but absent on those in area CA1 of the hippocampus. Here, we examined the effect of aging on synapses on projection neurons forming a hippocampal-cortico-thalamic circuit important for spatial working memory tasks from two genetically distinct mouse strains that exhibit susceptibility (C57BL/6J) or resistance (PWK/PhJ) to cognitive decline during aging. Across both strains, synapse density on CA1-to-PFC projection neurons appeared completely intact with age. In contrast, we found synapse loss on PFC-to-nucleus reuniens (RE) projection neurons from aged C57BL/6J but not PWK/PhJ mice. Moreover, synapses from aged PWK/PhJ mice but not from C57BL/6J exhibited altered morphologies that suggest increased efficiency to drive depolarization in the parent dendrite. Our findings suggest resistance to age-related cognitive decline results in part by age-related synaptic adaptations, and identification of these mechanisms in PWK/PhJ mice could uncover new therapeutic targets for promoting successful cognitive aging and extending human health span.


Subject(s)
Hippocampus , Neurons , Humans , Mice , Animals , Aged , Mice, Inbred C57BL , Hippocampus/physiology , Pyramidal Cells , Synapses/physiology , Neuronal Plasticity/genetics
5.
bioRxiv ; 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37546799

ABSTRACT

The disconnection of neuronal circuits through synaptic loss is presumed to be a major driver of age-related cognitive decline. Age-related cognitive decline is heterogeneous, yet whether genetic mechanisms differentiate successful from unsuccessful cognitive decline through synaptic structural mechanisms remains unknown. Previous work using rodent and primate models leveraged various techniques to suggest that age-related synaptic loss is widespread on pyramidal cells in prefrontal cortex (PFC) circuits but absent on those in area CA1 of the hippocampus. Here, we examined the effect of aging on synapses on projection neurons forming a hippocampal-cortico-thalamic circuit important for spatial working memory tasks from two genetically distinct mouse strains that exhibit susceptibility (C57BL/6J) or resistance (PWK/PhJ) to cognitive decline during aging. Across both strains, synapses on the CA1-to-PFC projection neurons appeared completely intact with age. In contrast, we found synapse loss on PFC-to-nucleus reuniens (RE) projection neurons from aged C57BL/6J but not PWK/PhJ mice. Moreover, synapses from aged PWK/PhJ mice but not from C57BL/6J exhibited morphological changes that suggest increased synaptic efficiency to depolarize the parent dendrite. Our findings suggest resistance to age-related cognitive decline results in part by age-related synaptic adaptations, and identification of these mechanisms in PWK/PhJ mice could uncover new therapeutic targets for promoting successful cognitive aging and extending human health span.

6.
bioRxiv ; 2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37162819

ABSTRACT

Common features of Alzheimer's disease (AD) include amyloid pathology, microglia activation and synaptic dysfunction, however, the causal relationships amongst them remains unclear. Further, human data suggest susceptibility and resilience to AD neuropathology is controlled by genetic context, a factor underexplored in mouse models. To this end, we leveraged viral strategies to label an AD-vulnerable neuronal circuit in CA1 dendrites projecting to the frontal cortex in genetically diverse C57BL/6J (B6) and PWK/PhJ (PWK) APP/PS1 mouse strains and used PLX5622 to non-invasively deplete brain microglia. Reconstructions of labeled neurons revealed microglia-dependent changes in dendritic spine density and morphology in B6 wild-type (WT) and APP/PS1 yet a marked stability of spines across PWK mice. We further showed that synaptic changes depend on direct microglia-dendrite interactions in B6. APP/PS1 but not PWK. APP/PS1 mice. Collectively, these results demonstrate that microglia-dependent synaptic alterations in a specific AD-vulnerable projection pathway are differentially controlled by genetic context.

7.
bioRxiv ; 2023 Mar 26.
Article in English | MEDLINE | ID: mdl-36993655

ABSTRACT

Neuronal mitochondria play important roles beyond ATP generation, including Ca2+ uptake, and therefore have instructive roles in synaptic function and neuronal response properties. Mitochondrial morphology differs significantly in the axon and dendrites of a given neuronal subtype, but in CA1 pyramidal neurons (PNs) of the hippocampus, mitochondria within the dendritic arbor also display a remarkable degree of subcellular, layer-specific compartmentalization. In the dendrites of these neurons, mitochondria morphology ranges from highly fused and elongated in the apical tuft, to more fragmented in the apical oblique and basal dendritic compartments, and thus occupy a smaller fraction of dendritic volume than in the apical tuft. However, the molecular mechanisms underlying this striking degree of subcellular compartmentalization of mitochondria morphology are unknown, precluding the assessment of its impact on neuronal function. Here, we demonstrate that this compartment-specific morphology of dendritic mitochondria requires activity-dependent, Camkk2-dependent activation of AMPK and its ability to phosphorylate two direct effectors: the pro-fission Drp1 receptor Mff and the recently identified anti-fusion, Opa1-inhibiting protein, Mtfr1l. Our study uncovers a new activity-dependent molecular mechanism underlying the extreme subcellular compartmentalization of mitochondrial morphology in dendrites of neurons in vivo through spatially precise regulation of mitochondria fission/fusion balance.

8.
Cell Rep ; 37(3): 109837, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686328

ABSTRACT

The selection of goal-directed behaviors is supported by neural circuits located within the frontal cortex. Frontal cortical afferents arise from multiple brain areas, yet the cell-type-specific targeting of these inputs is unclear. Here, we use monosynaptic retrograde rabies mapping to examine the distribution of afferent neurons targeting distinct classes of local inhibitory interneurons and excitatory projection neurons in mouse infralimbic frontal cortex. Interneurons expressing parvalbumin, somatostatin, or vasoactive intestinal peptide receive a large proportion of inputs from the hippocampus, while interneurons expressing neuron-derived neurotrophic factor receive a large proportion of inputs from thalamic regions. A similar dichotomy is present among the four different excitatory projection neurons. These results show a prominent bias among long-range hippocampal and thalamic afferent systems in their targeting to specific sets of frontal cortical neurons. Moreover, they suggest the presence of two distinct local microcircuits that control how different inputs govern frontal cortical information processing.


Subject(s)
Frontal Lobe/physiology , Hippocampus/physiology , Interneurons/physiology , Synapses/physiology , Thalamus/physiology , Animals , Behavior, Animal , Frontal Lobe/cytology , Frontal Lobe/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Interneurons/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neural Inhibition , Neural Pathways/cytology , Neural Pathways/metabolism , Neural Pathways/physiology , Neuroanatomical Tract-Tracing Techniques , Parvalbumins/genetics , Parvalbumins/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Synapses/metabolism , Thalamus/cytology , Thalamus/metabolism , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
9.
Front Neuroinform ; 13: 52, 2019.
Article in English | MEDLINE | ID: mdl-31427940

ABSTRACT

The functional features of neural circuits are determined by a combination of properties that range in scale from projections systems across the whole brain to molecular interactions at the synapse. The burgeoning field of neurocartography seeks to map these relevant features of brain structure-spanning a volume ∼20 orders of magnitude-to determine how neural circuits perform computations supporting cognitive function and complex behavior. Recent technological breakthroughs in tissue sample preparation, high-throughput electron microscopy imaging, and automated image analyses have produced the first visualizations of all synaptic connections between neurons of invertebrate model systems. However, the sheer size of the central nervous system in mammals implies that reconstruction of the first full brain maps at synaptic scale may not be feasible for decades. In this review, we outline existing and emerging technologies for neurocartography that complement electron microscopy-based strategies and are beginning to derive some basic organizing principles of circuit hodology at the mesoscale, microscale, and nanoscale. Specifically, we discuss how a host of light microscopy techniques including array tomography have been utilized to determine both long-range and subcellular organizing principles of synaptic connectivity. In addition, we discuss how new techniques, such as two-photon serial tomography of the entire mouse brain, have become attractive approaches to dissect the potential connectivity of defined cell types. Ultimately, principles derived from these techniques promise to facilitate a conceptual understanding of how connectomes, and neurocartography in general, can be effectively utilized toward reaching a mechanistic understanding of circuit function.

10.
Front Aging Neurosci ; 10: 146, 2018.
Article in English | MEDLINE | ID: mdl-29875653

ABSTRACT

Repeated exposure to stressors is known to produce large-scale remodeling of neurons within the prefrontal cortex (PFC). Recent work suggests stress-related forms of structural plasticity can interact with aging to drive distinct patterns of pyramidal cell morphological changes. However, little is known about how other cellular components within PFC might be affected by these challenges. Here, we examined the effects of stress exposure and aging on medial prefrontal cortical glial subpopulations. Interestingly, we found no changes in glial morphology with stress exposure but a profound morphological change with aging. Furthermore, we found an upregulation of non-nuclear glucocorticoid receptors (GR) with aging, while nuclear levels remained largely unaffected. Both changes are selective for microglia, with no stress or aging effect found in astrocytes. Lastly, we show that the changes found within microglia inversely correlated with the density of dendritic spines on layer III pyramidal cells. These findings suggest microglia play a selective role in synaptic health within the aging brain.

11.
Nat Neurosci ; 21(3): 353-363, 2018 03.
Article in English | MEDLINE | ID: mdl-29459763

ABSTRACT

CA1 pyramidal neurons are a major output of the hippocampus and encode features of experience that constitute episodic memories. Feature-selective firing of these neurons results from the dendritic integration of inputs from multiple brain regions. While it is known that synchronous activation of spatially clustered inputs can contribute to firing through the generation of dendritic spikes, there is no established mechanism for spatiotemporal synaptic clustering. Here we show that single presynaptic axons form multiple, spatially clustered inputs onto the distal, but not proximal, dendrites of CA1 pyramidal neurons. These compound connections exhibit ultrastructural features indicative of strong synapses and occur much more commonly in entorhinal than in thalamic afferents. Computational simulations revealed that compound connections depolarize dendrites in a biophysically efficient manner, owing to their inherent spatiotemporal clustering. Our results suggest that distinct afferent projections use different connectivity motifs that differentially contribute to dendritic integration.


Subject(s)
Axons/physiology , CA1 Region, Hippocampal/physiology , Dendrites/physiology , Pyramidal Cells/pathology , Synapses/physiology , Animals , Axons/ultrastructure , CA1 Region, Hippocampal/ultrastructure , Computer Simulation , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials , Male , Mice , Mice, Inbred C57BL , Neurons, Afferent/physiology , Neurons, Afferent/ultrastructure , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Pyramidal Cells/ultrastructure , Synapses/ultrastructure , Thalamus/cytology , Thalamus/physiology
12.
Neuron ; 89(5): 1016-30, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26898780

ABSTRACT

Neuronal circuit function is governed by precise patterns of connectivity between specialized groups of neurons. The diversity of GABAergic interneurons is a hallmark of cortical circuits, yet little is known about their targeting to individual postsynaptic dendrites. We examined synaptic connectivity between molecularly defined inhibitory interneurons and CA1 pyramidal cell dendrites using correlative light-electron microscopy and large-volume array tomography. We show that interneurons can be highly selective in their connectivity to specific dendritic branch types and, furthermore, exhibit precisely targeted connectivity to the origin or end of individual branches. Computational simulations indicate that the observed subcellular targeting enables control over the nonlinear integration of synaptic input or the initiation and backpropagation of action potentials in a branch-selective manner. Our results demonstrate that connectivity between interneurons and pyramidal cell dendrites is more precise and spatially segregated than previously appreciated, which may be a critical determinant of how inhibition shapes dendritic computation.


Subject(s)
CA1 Region, Hippocampal/cytology , Dendrites/physiology , Neural Inhibition/physiology , Neurons/cytology , Neurons/physiology , Action Potentials/drug effects , Action Potentials/genetics , Action Potentials/physiology , Animals , Computer Simulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Models, Neurological , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Parvalbumins/genetics , Parvalbumins/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Synapses/metabolism , Synapses/physiology , Synapses/ultrastructure , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
13.
Neuron ; 87(6): 1274-1289, 2015 Sep 23.
Article in English | MEDLINE | ID: mdl-26402609

ABSTRACT

Spatial and temporal features of synaptic inputs engage integration mechanisms on multiple scales, including presynaptic release sites, postsynaptic dendrites, and networks of inhibitory interneurons. Here we investigate how these mechanisms cooperate to filter synaptic input in hippocampal area CA1. Dendritic recordings from CA1 pyramidal neurons reveal that proximal inputs from CA3 as well as distal inputs from entorhinal cortex layer III (ECIII) sum sublinearly or linearly at low firing rates due to feedforward inhibition, but sum supralinearly at high firing rates due to synaptic facilitation, producing a high-pass filter. However, during ECIII and CA3 input comparison, supralinear dendritic integration is dynamically balanced by feedforward and feedback inhibition, resulting in suppression of dendritic complex spiking. We find that a particular subpopulation of CA1 interneurons expressing neuropeptide Y (NPY) contributes prominently to this dynamic filter by integrating both ECIII and CA3 input pathways and potently inhibiting CA1 pyramidal neuron dendrites.


Subject(s)
Action Potentials/physiology , CA1 Region, Hippocampal/physiology , Interneurons/physiology , Nerve Net/physiology , Neural Inhibition/physiology , Animals , Gene Knock-In Techniques/methods , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Rats
14.
Nat Methods ; 12(6): 568-76, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25915120

ABSTRACT

We describe an engineered family of highly antigenic molecules based on GFP-like fluorescent proteins. These molecules contain numerous copies of peptide epitopes and simultaneously bind IgG antibodies at each location. These 'spaghetti monster' fluorescent proteins (smFPs) distributed well in neurons, notably into small dendrites, spines and axons. smFP immunolabeling localized weakly expressed proteins not well resolved with traditional epitope tags. By varying epitope and scaffold, we generated a diverse family of mutually orthogonal antigens. In cultured neurons and mouse and fly brains, smFP probes allowed robust, orthogonal multicolor visualization of proteins, cell populations and neuropil. smFP variants complement existing tracers and greatly increase the number of simultaneous imaging channels, and they performed well in advanced preparations such as array tomography, super-resolution fluorescence imaging and electron microscopy. In living cells, the probes improved single-molecule image tracking and increased yield for RNA-seq. These probes facilitate new experiments in connectomics, transcriptomics and protein localization.


Subject(s)
Luminescent Proteins/chemistry , Microscopy, Electron/methods , Microscopy, Fluorescence/methods , Animals , Antigens , Brain Mapping , Drosophila , Mice , Models, Molecular , Molecular Sequence Data , Neurons , Protein Conformation
15.
Exp Neurol ; 241: 5-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23219884

ABSTRACT

Chronic immobilization stress (CIS) shortens apical dendritic trees of CA3 pyramidal neurons in the hippocampus of the male rat, and dendritic length may be a determinant of vulnerability to stress. Expression of the polysialylated form of neural cell adhesion molecule (PSA-NCAM) in the hippocampal formation is increased by stress, while PSA removal by Endo-neuraminidase-N (endo-N) is known to cause the mossy fibers to defasciculate and synapse ectopically in their CA3 target area. We show here that enzymatic removal of PSA produced a remarkable expansion of dendritic arbors of CA3 pyramidal neurons, with a lesser effect in CA1. This expansion eclipsed the CIS-induced shortening of CA3 dendrites, with the expanded dendrites of both no-stress-endo-N and CIS-endo-N rats being longer than those in no-stress-control rats and much longer than those in CIS-control rats. As predicted by the hypothesis that endo-N-induced dendritic expansion might increase vulnerability to excitotoxic challenge, systemic injection with kainic acid, showed markedly increased neuronal degeneration, as assessed by fluorojade B histochemistry, in rats that had been treated with endo-N compared to vehicle-treated rats throughout the entire hippocampal formation. PSA removal also exacerbated the CIS-induced reduction in body weight and abolished effects of CIS on NPY and NR2B mRNA levels. These findings support the hypothesis that CA3 arbor plasticity plays a protective role during prolonged stress and clarify the role of PSA-NCAM in stress-induced dendritic plasticity.


Subject(s)
CA3 Region, Hippocampal/pathology , Dendrites/pathology , Neural Cell Adhesion Molecules/metabolism , Pyramidal Cells/pathology , Sialic Acids/deficiency , Stress, Psychological/pathology , Analysis of Variance , Animals , Body Mass Index , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , Dendrites/drug effects , Dendrites/ultrastructure , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Fluoresceins , Gene Expression Regulation/drug effects , Kainic Acid/toxicity , Male , Metalloendopeptidases/pharmacology , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Neural Cell Adhesion Molecules/drug effects , Organic Chemicals , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Silver Staining , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
16.
Neurobiol Aging ; 34(1): 200-10, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22727942

ABSTRACT

Age-related impairments of executive functions appear to be related to reductions of the number and plasticity of dendritic spine synapses in the prefrontal cortex (PFC). Experimental evidence suggests that synaptic plasticity is mediated by the spine actin cytoskeleton, and a major pathway regulating actin-based plasticity is controlled by phosphorylated LIM kinase (pLIMK). We asked whether aging resulted in altered synaptic density, morphology, and pLIMK expression in the rat prelimbic region of the PFC. Using unbiased electron microscopy, we found an approximate 50% decrease in the density of small synapses with aging, while the density of large synapses remained unchanged. Postembedding immunogold revealed that pLIMK localized predominantly to the postsynaptic density where it was increased in aging synapses by approximately 50%. Furthermore, the age-related increase in pLIMK occurred selectively within the largest subset of prelimbic PFC synapses. Because pLIMK is known to inhibit actin filament plasticity, these data support the hypothesis that age-related increases in pLIMK may explain the stability of large synapses at the expense of their plasticity.


Subject(s)
Aging , Lim Kinases/metabolism , Neurons/cytology , Prefrontal Cortex/cytology , Synapses/ultrastructure , Age Factors , Analysis of Variance , Animals , Gene Expression Regulation , Male , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Post-Synaptic Density/metabolism , Post-Synaptic Density/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/metabolism
17.
Neuron ; 76(4): 776-89, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23177962

ABSTRACT

Relating the function of neuronal cell types to information processing and behavior is a central goal of neuroscience. In the hippocampus, pyramidal cells in CA1 and the subiculum process sensory and motor cues to form a cognitive map encoding spatial, contextual, and emotional information, which they transmit throughout the brain. Do these cells constitute a single class or are there multiple cell types with specialized functions? Using unbiased cluster analysis, we show that there are two morphologically and electrophysiologically distinct principal cell types that carry hippocampal output. We show further that these two cell types are inversely modulated by the synergistic action of glutamate and acetylcholine acting on metabotropic receptors that are central to hippocampal function. Combined with prior connectivity studies, our results support a model of hippocampal processing in which the two pyramidal cell types are predominantly segregated into two parallel pathways that process distinct modalities of information.


Subject(s)
CA1 Region, Hippocampal/cytology , Neurons/classification , Neurons/physiology , Receptors, Metabotropic Glutamate/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Benzylamines/pharmacology , Biophysics , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Transporter 3/metabolism , GABA Antagonists/pharmacology , Glutamate Decarboxylase/metabolism , In Vitro Techniques , Lysine/analogs & derivatives , Lysine/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Patch-Clamp Techniques , Phosphinic Acids/pharmacology , Pyridazines/pharmacology , Rats , Time Factors
18.
J Neurosci ; 32(34): 11700-5, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22915112

ABSTRACT

Preclinical animal models have provided strong evidence that estrogen (E) therapy (ET) enhances cognition and induces spinogenesis in neuronal circuits. However, clinical studies have been inconsistent, with some studies revealing adverse effects of ET, including an increased risk of dementia. In an effort to bridge this disconnect between the preclinical and clinical data, we have developed a nonhuman primate (NHP) model of ET combined with high-resolution dendritic spine analysis of dorsolateral prefrontal cortical (dlPFC) neurons. Previously, we reported cyclic ET in aged, ovariectomized NHPs increased spine density on dlPFC neurons. Here, we report that monkeys treated with cyclic E treatment paired with cyclic progesterone (P), continuous E combined with P (either cyclic or continuous), or unopposed continuous E failed to increase spines on dlPFC neurons. Given that the most prevalent form of ET prescribed to women is a combined and continuous E and P, these data bring into convergence the human neuropsychological findings and preclinical neurobiological evidence that standard hormone therapy in women is unlikely to yield the synaptic benefit presumed to underlie the cognitive enhancement reported in animal models.


Subject(s)
Aging/drug effects , Dendritic Spines/drug effects , Estrogens/pharmacology , Neurons/cytology , Prefrontal Cortex/cytology , Progesterone/pharmacology , Aging/pathology , Analysis of Variance , Animals , Estrogens/blood , Female , Macaca mulatta , Microscopy, Confocal , Neurons/drug effects , Ovariectomy , Prefrontal Cortex/drug effects , Progesterone/blood
19.
J Neurosci ; 31(21): 7831-9, 2011 May 25.
Article in English | MEDLINE | ID: mdl-21613496

ABSTRACT

Cognitive functions that require the prefrontal cortex are highly sensitive to aging in humans, nonhuman primates, and rodents, although the neurobiological correlates of this vulnerability remain largely unknown. It has been proposed that dendritic spines represent the primary site of structural plasticity in the adult brain, and recent data have supported the hypothesis that aging is associated with alterations of dendritic spine morphology and plasticity in prefrontal cortex. However, no study to date has directly examined whether aging alters the capacity for experience-dependent spine plasticity in aging prefrontal neurons. To address this possibility, we used young, middle-aged, and aged rats in a behavioral stress paradigm known to produce spine remodeling in prefrontal cortical neurons. In young rats, stress resulted in dendritic spine loss and altered patterns of spine morphology; in contrast, spines from middle-aged and aged animals were remarkably stable and did not show evidence of remodeling. The loss of stress-induced spine plasticity observed in aging rats occurred alongside robust age-related reductions in spine density and shifts in remaining spine morphology. Together, the data presented here provide the first evidence that experience-dependent spine plasticity is altered by aging in prefrontal cortex, and support a model in which dendritic spines become progressively less plastic in the aging brain.


Subject(s)
Aging/physiology , Dendritic Spines/physiology , Learning/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Animals , Cognition/physiology , Dendritic Spines/ultrastructure , Male , Prefrontal Cortex/ultrastructure , Rats , Rats, Sprague-Dawley
20.
Proc Natl Acad Sci U S A ; 108(4): 1657-62, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21220317

ABSTRACT

Circadian (daily) rhythms are present in almost all plants and animals. In mammals, a brain clock located in the hypothalamic suprachiasmatic nucleus maintains synchrony between environmental light/dark cycles and physiology and behavior. Over the past 100 y, especially with the advent of electric lighting, modern society has resulted in a round-the-clock lifestyle, in which natural connections between rest/activity cycles and environmental light/dark cycles have been degraded or even broken. Instances in which rapid changes to sleep patterns are necessary, such as transmeridian air travel, demonstrate negative effects of acute circadian disruption on physiology and behavior. However, the ramifications of chronic disruption of the circadian clock for mental and physical health are not yet fully understood. By housing mice in 20-h light/dark cycles, incongruous with their endogenous ∼24-h circadian period, we were able to model the effects of chronic circadian disruption noninvasively. Housing in these conditions results in accelerated weight gain and obesity, as well as changes in metabolic hormones. In the brain, circadian-disrupted mice exhibit a loss of dendritic length and decreased complexity of neurons in the prelimbic prefrontal cortex, a brain region important in executive function and emotional control. Disrupted animals show decreases in cognitive flexibility and changes in emotionality consistent with the changes seen in neural architecture. How our findings translate to humans living and working in chronic circadian disruption is unknown, but we believe that this model can provide a foundation to understand how environmental disruption of circadian rhythms impacts the brain, behavior, and physiology.


Subject(s)
Behavior, Animal/physiology , Brain/physiology , Circadian Clocks/physiology , Energy Metabolism/physiology , Animals , Biological Clocks/physiology , Blood Glucose/metabolism , Body Temperature/physiology , Brain/cytology , Circadian Rhythm/physiology , Humans , Insulin/blood , Leptin/blood , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Neurons/physiology , Photoperiod , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Weight Gain/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...