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










Publication year range
1.
Brain Res ; 1841: 149083, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38866308

ABSTRACT

Alzheimer's disease (AD) affects both grey and white matter (WM), but considerably more is known about the former. Interestingly, WM disruption has been consistently observed and thoroughly described using imaging modalities, particularly MRI which has shown WM functional disconnections between the hippocampus and other brain regions during AD pathogenesis when early neurodegeneration and synapse loss are also evident. Nonetheless, high-resolution structural and functional analyses of WM during AD pathogenesis remain scarce. Given the importance of the myelinated axons in the WM for conveying information across brain regions, such studies will provide valuable information on the cellular drivers and consequences of WM disruption that contribute to the characteristic cognitive decline of AD. Here, we employed a multi-scale approach to investigate hippocampal WM disruption during AD pathogenesis and determine whether hippocampal WM changes accompany the well-documented grey matter losses. Our data indicate that ultrastructural myelin disruption is elevated in the alveus in human AD cases and increases with age in 5xFAD mice. Unreliable action potential propagation and changes to sodium channel expression at the node of Ranvier co-emerged with this deterioration. These findings provide important insight to the neurobiological substrates and functional consequences of decreased WM integrity and are consistent with the notion that hippocampal disconnection contributes to cognitive changes in AD.

2.
J Mol Cell Biol ; 2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37682518

ABSTRACT

Synaptic vesicles can undergo several modes of exocytosis, endocytosis, and trafficking within individual synapses, and their fates may be linked to differences in the vesicular protein composition. Here, we mapped the intrasynaptic distribution of the synaptic vesicle proteins SV2B and SV2A in glutamatergic synapses of the hippocampus using three-dimensional electron microscopy. SV2B is almost completely absent from both docked vesicles and a distinct cluster of vesicles found near the active zone. In contrast, SV2A was found in all domains of the synapse and was slightly enriched near the active zone. SV2B and SV2A were found on the membrane in the peri-active zone, suggesting recycling from both clusters of vesicles. SV2B knockout mice displayed an increased seizure induction threshold only in a model employing high-frequency stimulation. Our data show that glutamatergic synapses generate molecularly distinct populations of synaptic vesicles and are able to maintain them at steep spatial gradients. The almost complete absence of SV2B from vesicles at the active zone of wildtype mice may explain why SV2A has been found to be more important for vesicle release.

3.
Neurobiol Aging ; 106: 207-222, 2021 10.
Article in English | MEDLINE | ID: mdl-34303222

ABSTRACT

The hippocampus is vulnerable to deterioration in Alzheimer's disease (AD). It is, however, a heterogeneous structure, which may contribute to the differential volumetric changes along its septotemporal axis during AD progression. Here, we investigated amyloid plaque deposition along the dorsoventral axis in two strains of transgenic AD (ADTg) mouse models. We also used patch-clamp physiology in these mice to probe for functional consequences of AD pathogenesis in ventral hippocampus, which we found bears significantly higher plaque burden in the aged ADTg group compared to corresponding dorsal regions. Despite dorsoventral differences in amyloid load, ventral CA1 pyramidal neurons of aged ADTg mice exhibited subthreshold physiological changes similar to those previously reported in dorsal neurons, indicative of an HCN channelopathy, but lacked exacerbated suprathreshold accommodation. Additionally, HCN channel function could be rescued by pharmacological manipulation of the endoplasmic reticulum. These observations suggest that an AD-linked HCN channelopathy emerges in both dorsal and ventral CA1 pyramidal neurons, but that the former encounter an additional integrative obstacle in the form of reduced intrinsic excitability.


Subject(s)
Aging/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Plaque, Amyloid/metabolism , Pyramidal Cells/metabolism , Signal Transduction , Animals , Disease Models, Animal , Disease Progression , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Mice, Transgenic , Organ Size , Patch-Clamp Techniques
4.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33593893

ABSTRACT

Behaviors that rely on the hippocampus are particularly susceptible to chronological aging, with many aged animals (including humans) maintaining cognition at a young adult-like level, but many others the same age showing marked impairments. It is unclear whether the ability to maintain cognition over time is attributable to brain maintenance, sufficient cognitive reserve, compensatory changes in network function, or some combination thereof. While network dysfunction within the hippocampal circuit of aged, learning-impaired animals is well-documented, its neurobiological substrates remain elusive. Here we show that the synaptic architecture of hippocampal regions CA1 and CA3 is maintained in a young adult-like state in aged rats that performed comparably to their young adult counterparts in both trace eyeblink conditioning and Morris water maze learning. In contrast, among learning-impaired, but equally aged rats, we found that a redistribution of synaptic weights amplifies the influence of autoassociational connections among CA3 pyramidal neurons, yet reduces the synaptic input onto these same neurons from the dentate gyrus. Notably, synapses within hippocampal region CA1 showed no group differences regardless of cognitive ability. Taking the data together, we find the imbalanced synaptic weights within hippocampal CA3 provide a substrate that can explain the abnormal firing characteristics of both CA3 and CA1 pyramidal neurons in aged, learning-impaired rats. Furthermore, our work provides some clarity with regard to how some animals cognitively age successfully, while others' lifespans outlast their "mindspans."


Subject(s)
CA1 Region, Hippocampal/pathology , CA3 Region, Hippocampal/pathology , Cognitive Aging , Pyramidal Cells/pathology , Synapses/pathology , Animals , Male , Rats , Rats, Inbred BN , Rats, Inbred F344
5.
Cell Rep ; 30(10): 3520-3535.e7, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32160554

ABSTRACT

BIN1, a member of the BAR adaptor protein family, is a significant late-onset Alzheimer disease risk factor. Here, we investigate BIN1 function in the brain using conditional knockout (cKO) models. Loss of neuronal Bin1 expression results in the select impairment of spatial learning and memory. Examination of hippocampal CA1 excitatory synapses reveals a deficit in presynaptic release probability and slower depletion of neurotransmitters during repetitive stimulation, suggesting altered vesicle dynamics in Bin1 cKO mice. Super-resolution and immunoelectron microscopy localizes BIN1 to presynaptic sites in excitatory synapses. Bin1 cKO significantly reduces synapse density and alters presynaptic active zone protein cluster formation. Finally, 3D electron microscopy reconstruction analysis uncovers a significant increase in docked and reserve pools of synaptic vesicles at hippocampal synapses in Bin1 cKO mice. Our results demonstrate a non-redundant role for BIN1 in presynaptic regulation, thus providing significant insights into the fundamental function of BIN1 in synaptic physiology relevant to Alzheimer disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Memory Consolidation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Brain/metabolism , Excitatory Postsynaptic Potentials , Mice, Inbred C57BL , Mice, Knockout , Neurons/ultrastructure , Presynaptic Terminals/ultrastructure , Recognition, Psychology , SNARE Proteins/metabolism , Spatial Learning
6.
Brain Pathol ; 29(4): 485-501, 2019 07.
Article in English | MEDLINE | ID: mdl-30506549

ABSTRACT

Bridging integrator 1 (BIN1) is the most significant late-onset Alzheimer's disease (AD) susceptibility locus identified via genome-wide association studies. BIN1 is an adaptor protein that regulates membrane dynamics in the context of endocytosis and membrane remodeling. An increase in BIN1 expression and changes in the relative levels of alternatively spliced BIN1 isoforms have been reported in the brains of patients with AD. BIN1 can bind to Tau, and an increase in BIN1 expression correlates with Tau pathology. In contrast, the loss of BIN1 expression in cultured cells elevates Aß production and Tau propagation by insfluencing endocytosis and recycling. Here, we show that BIN1 accumulates adjacent to amyloid deposits in vivo. We found an increase in insoluble BIN1 and a striking accrual of BIN1 within and near amyloid deposits in the brains of multiple transgenic models of AD. The peri-deposit aberrant BIN1 localization was conspicuously different from the accumulation of APP and BACE1 within dystrophic neurites. Although BIN1 is highly expressed in mature oligodendrocytes, BIN1 association with amyloid deposits occurred in the absence of the accretion of other oligodendrocyte or myelin proteins. Finally, super-resolution microscopy and immunogold electron microscopy analyses highlight the presence of BIN1 in proximity to amyloid fibrils at the edges of amyloid deposits. These results reveal the aberrant accumulation of BIN1 is a feature associated with AD amyloid pathology. Our findings suggest a potential role for BIN1 in extracellular Aß deposition in vivo that is distinct from its well-characterized function as an adaptor protein in endocytosis and membrane remodeling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/pathology , Nuclear Proteins/metabolism , Plaque, Amyloid/pathology , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/physiology , Alzheimer Disease/metabolism , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/pathology , Animals , Brain/pathology , Disease Models, Animal , Female , Genome-Wide Association Study , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/physiology , Nuclear Proteins/physiology , Plaque, Amyloid/metabolism , Signal Transduction , Tumor Suppressor Proteins/physiology , tau Proteins/metabolism
7.
Neurobiol Learn Mem ; 154: 141-157, 2018 10.
Article in English | MEDLINE | ID: mdl-29906573

ABSTRACT

Voltage-gated ion channels are critical for neuronal integration. Some of these channels, however, are misregulated in several neurological disorders, causing both gain- and loss-of-function channelopathies in neurons. Using several transgenic mouse models of Alzheimer's disease (AD), we find that sub-threshold voltage signals strongly influenced by hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels progressively deteriorate over chronological aging in hippocampal CA1 pyramidal neurons. The degraded signaling via HCN channels in the transgenic mice is accompanied by an age-related global loss of their non-uniform dendritic expression. Both the aberrant signaling via HCN channels and their mislocalization could be restored using a variety of pharmacological agents that target the endoplasmic reticulum (ER). Our rescue of the HCN channelopathy helps provide molecular details into the favorable outcomes of ER-targeting drugs on the pathogenesis and synaptic/cognitive deficits in AD mouse models, and implies that they might have beneficial effects on neurological disorders linked to HCN channelopathies.


Subject(s)
Alzheimer Disease/physiopathology , CA1 Region, Hippocampal/physiology , Channelopathies/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Neuronal Plasticity , Pyramidal Cells/physiology , Action Potentials , Aging , Animals , CA1 Region, Hippocampal/ultrastructure , Disease Models, Animal , Endoplasmic Reticulum/physiology , Female , Male , Mice, Transgenic , Pyramidal Cells/ultrastructure
8.
J Neurosci ; 37(47): 11298-11310, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29038238

ABSTRACT

Fragile X syndrome (FXS) is a neurodevelopmental disorder that is a leading cause of inherited intellectual disability, and the most common known cause of autism spectrum disorder. FXS is broadly characterized by sensory hypersensitivity and several developmental alterations in synaptic and circuit function have been uncovered in the sensory cortex of the mouse model of FXS (Fmr1 KO). GABA-mediated neurotransmission and fast-spiking (FS) GABAergic interneurons are central to cortical circuit development in the neonate. Here we demonstrate that there is a delay in the maturation of the intrinsic properties of FS interneurons in the sensory cortex, and a deficit in the formation of excitatory synaptic inputs on to these neurons in neonatal Fmr1 KO mice. Both these delays in neuronal and synaptic maturation were rectified by chronic administration of a TrkB receptor agonist. These results demonstrate that the maturation of the GABAergic circuit in the sensory cortex is altered during a critical developmental period due in part to a perturbation in BDNF-TrkB signaling, and could contribute to the alterations in cortical development underlying the sensory pathophysiology of FXS.SIGNIFICANCE STATEMENT Fragile X (FXS) individuals have a range of sensory related phenotypes, and there is growing evidence of alterations in neuronal circuits in the sensory cortex of the mouse model of FXS (Fmr1 KO). GABAergic interneurons are central to the correct formation of circuits during cortical critical periods. Here we demonstrate a delay in the maturation of the properties and synaptic connectivity of interneurons in Fmr1 KO mice during a critical period of cortical development. The delays both in cellular and synaptic maturation were rectified by administration of a TrkB receptor agonist, suggesting reduced BDNF-TrkB signaling as a contributing factor. These results provide evidence that the function of fast-spiking interneurons is disrupted due to a deficiency in neurotrophin signaling during early development in FXS.


Subject(s)
Excitatory Postsynaptic Potentials , Fragile X Syndrome/metabolism , GABAergic Neurons/metabolism , Interneurons/metabolism , Receptor, trkB/metabolism , Animals , Female , Fragile X Mental Retardation Protein/genetics , GABAergic Neurons/cytology , GABAergic Neurons/physiology , Interneurons/cytology , Interneurons/physiology , Mice , Mice, Inbred C57BL , Receptor, trkB/agonists , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiology
9.
Ann Neurol ; 81(1): 46-57, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27900791

ABSTRACT

OBJECTIVE: The main goal of dopamine cell replacement therapy in Parkinson disease (PD) is to provide clinical benefit mediated by graft survival with nigrostriatal reinnervation. We report a dichotomy between graft structure and clinical function in a patient dying 16 years following fetal nigral grafting. METHODS: A 55-year-old levodopa-responsive woman with PD received bilateral putaminal fetal mesencephalic grafts as part of an NIH-sponsored double-blind sham-controlled trial. The patient never experienced clinical benefit, and her course was complicated by the development of graft-related dyskinesias. Fluorodopa positron emission tomography demonstrated significant increases postgrafting bilaterally. She experienced worsening of parkinsonism with severe dyskinesias, and underwent subthalamic nucleus deep brain stimulation 8 years after grafting. She died 16 years after transplantation. RESULTS: Postmortem analyses confirmed the diagnosis of PD and demonstrated >300,000 tyrosine hydroxylase (TH)-positive grafted cells per side with normalized striatal TH-immunoreactive fiber innervation and bidirectional synaptic connectivity. Twenty-seven percent and 17% of grafted neurons were serine 129-phosphorylated α-synuclein positive in the left and right putamen, respectively. INTERPRETATION: These findings represent the largest number of surviving dopamine neurons and the densest and most widespread graft-mediated striatal dopamine reinnervation following a transplant procedure reported to date. Despite this, clinical recovery was not observed. Furthermore, the grafts were associated with a form of dyskinesias that resembled diphasic dyskinesia and persisted in the off-medication state. We hypothesize that the grafted cells produced a low level of dopamine sufficient to cause a levodopa-independent continuous form of diphasic dyskinesias, but insufficient to provide an antiparkinsonian benefit. ANN NEUROL 2017;81:46-57.


Subject(s)
Dopaminergic Neurons/metabolism , Graft Survival , Mesencephalon/transplantation , Parkinson Disease/surgery , Brain Tissue Transplantation , Dopaminergic Neurons/ultrastructure , Female , Humans , Middle Aged , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
10.
J Neurosci ; 35(38): 13206-18, 2015 Sep 23.
Article in English | MEDLINE | ID: mdl-26400949

ABSTRACT

Aging-related impairments in hippocampus-dependent cognition have been attributed to maladaptive changes in the functional properties of pyramidal neurons within the hippocampal subregions. Much evidence has come from work on CA1 pyramidal neurons, with CA3 pyramidal neurons receiving comparatively less attention despite its age-related hyperactivation being postulated to interfere with spatial processing in the hippocampal circuit. Here, we use whole-cell current-clamp to demonstrate that aged rat (29-32 months) CA3 pyramidal neurons fire significantly more action potentials (APs) during theta-burst frequency stimulation and that this is associated with faster AP repolarization (i.e., narrower AP half-widths and enlarged fast afterhyperpolarization). Using a combination of patch-clamp physiology, pharmacology, Western blot analyses, immunohistochemistry, and array tomography, we demonstrate that these faster AP kinetics are mediated by enhanced function and expression of Kv4.2/Kv4.3 A-type K(+) channels, particularly within the perisomatic compartment, of CA3 pyramidal neurons. Thus, our study indicates that inhibition of these A-type K(+) channels can restore the intrinsic excitability properties of aged CA3 pyramidal neurons to a young-like state. Significance statement: Age-related learning deficits have been attributed, in part, to altered hippocampal pyramidal neuronal function with normal aging. Much evidence has come from work on CA1 neurons, with CA3 neurons receiving comparatively less attention despite its age-related hyperactivation being postulated to interfere with spatial processing. Hence, we conducted a series of experiments to identify the cellular mechanisms that underlie the hyperexcitability reported in the CA3 region. Contrary to CA1 neurons, we demonstrate that postburst afterhyperpolarization is not altered with aging and that aged CA3 pyramidal neurons are able to fire significantly more action potentials and that this is associated with faster action potential repolarization through enhanced expression of Kv4.2/Kv4.3 A-type K(+) channels, particularly within the cell bodies of CA3 pyramidal neurons.


Subject(s)
Aging/physiology , CA3 Region, Hippocampal/cytology , Membrane Potentials/physiology , Pyramidal Cells/physiology , Shal Potassium Channels/metabolism , Analysis of Variance , Animals , Biophysics , Dose-Response Relationship, Drug , Electric Stimulation , In Vitro Techniques , Male , Membrane Potentials/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Pyramidal Cells/drug effects , Rats , Rats, Inbred F344 , Synaptic Potentials/drug effects , Synaptic Potentials/physiology
11.
Brain Struct Funct ; 220(6): 3143-65, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25031178

ABSTRACT

Alzheimer's disease (AD) is associated with alterations in the distribution, number, and size of inputs to hippocampal neurons. Some of these changes are thought to be neurodegenerative, whereas others are conceptualized as compensatory, plasticity-like responses, wherein the remaining inputs reactively innervate vulnerable dendritic regions. Here, we provide evidence that the axospinous synapses of human AD cases and mice harboring AD-linked genetic mutations (the 5XFAD line) exhibit both, in the form of synapse loss and compensatory changes in the synapses that remain. Using array tomography, quantitative conventional electron microscopy, immunogold electron microscopy for AMPARs, and whole-cell patch-clamp physiology, we find that hippocampal CA1 pyramidal neurons in transgenic mice are host to an age-related synapse loss in their distal dendrites, and that the remaining synapses express more AMPA-type glutamate receptors. Moreover, the number of axonal boutons that synapse with multiple spines is significantly reduced in the transgenic mice. Through serial section electron microscopic analyses of human hippocampal tissue, we further show that putative compensatory changes in synapse strength are also detectable in axospinous synapses of proximal and distal dendrites in human AD cases, and that their multiple synapse boutons may be more powerful than those in non-cognitively impaired human cases. Such findings are consistent with the notion that the pathophysiology of AD is a multivariate product of both neurodegenerative and neuroplastic processes, which may produce adaptive and/or maladaptive responses in hippocampal synaptic strength and plasticity.


Subject(s)
Alzheimer Disease/pathology , CA1 Region, Hippocampal/pathology , Dendrites/pathology , Neurons/pathology , Pyramidal Cells/pathology , Alzheimer Disease/metabolism , Animals , Axons/metabolism , CA1 Region, Hippocampal/metabolism , Cells, Cultured , Dendrites/metabolism , Humans , Male , Mice , Mice, Transgenic , Models, Animal , Neuronal Plasticity , Neurons/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Pyramidal Cells/metabolism , Receptors, AMPA/metabolism , Synapses/pathology
12.
Neuron ; 80(6): 1451-63, 2013 Dec 18.
Article in English | MEDLINE | ID: mdl-24360547

ABSTRACT

Neuronal computation involves the integration of synaptic inputs that are often distributed over expansive dendritic trees, suggesting the need for compensatory mechanisms that enable spatially disparate synapses to influence neuronal output. In hippocampal CA1 pyramidal neurons, such mechanisms have indeed been reported, which normalize either the ability of distributed synapses to drive action potential initiation in the axon or their ability to drive dendritic spiking locally. Here we report that these mechanisms can coexist, through an elegant combination of distance-dependent regulation of synapse number and synaptic expression of AMPA and NMDA receptors. Together, these complementary gradients allow individual dendrites in both the apical and basal dendritic trees of hippocampal neurons to operate as facile computational subunits capable of supporting both global integration in the soma/axon and local integration in the dendrite.


Subject(s)
CA1 Region, Hippocampal/metabolism , Dendrites/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , CA1 Region, Hippocampal/physiology , CA1 Region, Hippocampal/ultrastructure , Dendrites/physiology , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials/physiology , Male , Membrane Potentials/physiology , Models, Neurological , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Pyramidal Cells/ultrastructure , Rats , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...