Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Semin Cell Dev Biol ; 139: 35-54, 2023 04.
Article in English | MEDLINE | ID: mdl-35963663

ABSTRACT

The early stages of Alzheimer's disease (AD) have been linked to microcircuit dysfunction and pathophysiological neuronal firing in several brain regions. Inhibitory GABAergic microcircuitry is a critical feature of stable neural-circuit function in the healthy brain, and its dysregulation has therefore been proposed as contributing to AD-related pathophysiology. However, exactly how the critical balance between excitatory and inhibitory microcircuitry is modified by AD pathogenesis remains unclear. Here, we set the current evidence implicating dysfunctional GABAergic microcircuitry as a driver of early AD pathophysiology in a simple conceptual framework. Our framework is based on a generalised reductionist model of firing-rate control by local feedback inhibition. We use this framework to consider multiple loci that may be vulnerable to disruption by AD pathogenesis. We first start with evidence investigating how AD-related processes may impact the gross number of inhibitory neurons in the network. We then move to discuss how pathology may impact intrinsic cellular properties and firing thresholds of GABAergic neurons. Finally, we cover how AD-related pathogenesis may disrupt synaptic connectivity between excitatory and inhibitory neurons. We use the feedback inhibition framework to discuss and organise the available evidence from both preclinical rodent work and human studies in AD patients and conclude by identifying key questions and understudied areas for future investigation.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/genetics , GABAergic Neurons , Brain
2.
Proc Natl Acad Sci U S A ; 119(43): e2206083119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36269859

ABSTRACT

Genome-wide association studies (GWASs) have identified genetic loci associated with the risk of Alzheimer's disease (AD), but the molecular mechanisms by which they confer risk are largely unknown. We conducted a metabolome-wide association study (MWAS) of AD-associated loci from GWASs using untargeted metabolic profiling (metabolomics) by ultraperformance liquid chromatography-mass spectrometry (UPLC-MS). We identified an association of lactosylceramides (LacCer) with AD-related single-nucleotide polymorphisms (SNPs) in ABCA7 (P = 5.0 × 10-5 to 1.3 × 10-44). We showed that plasma LacCer concentrations are associated with cognitive performance and genetically modified levels of LacCer are associated with AD risk. We then showed that concentrations of sphingomyelins, ceramides, and hexosylceramides were altered in brain tissue from Abca7 knockout mice, compared with wild type (WT) (P = 0.049-1.4 × 10-5), but not in a mouse model of amyloidosis. Furthermore, activation of microglia increases intracellular concentrations of hexosylceramides in part through induction in the expression of sphingosine kinase, an enzyme with a high control coefficient for sphingolipid and ceramide synthesis. Our work suggests that the risk for AD arising from functional variations in ABCA7 is mediated at least in part through ceramides. Modulation of their metabolism or downstream signaling may offer new therapeutic opportunities for AD.


Subject(s)
ATP-Binding Cassette Transporters , Alzheimer Disease , Ceramides , Animals , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Ceramides/metabolism , Chromatography, Liquid , Genome-Wide Association Study , Lactosylceramides , Metabolome , Mice, Knockout , Sphingomyelins , Tandem Mass Spectrometry
3.
Neurobiol Dis ; 135: 104744, 2020 02.
Article in English | MEDLINE | ID: mdl-31931139

ABSTRACT

Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation.


Subject(s)
Huntington Disease/microbiology , Myelin Proteins/metabolism , Myelin Sheath/pathology , White Matter/microbiology , Animals , Bacteria/isolation & purification , Corpus Callosum/metabolism , Corpus Callosum/microbiology , Disease Models, Animal , Huntington Disease/pathology , Mice, Transgenic , Myelin Sheath/metabolism , Neuronal Plasticity/physiology , Oligodendroglia/metabolism , Prefrontal Cortex/metabolism , White Matter/pathology
4.
Neurobiol Dis ; 127: 65-75, 2019 07.
Article in English | MEDLINE | ID: mdl-30802499

ABSTRACT

Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation.


Subject(s)
Corpus Callosum/pathology , Gastrointestinal Microbiome/physiology , Huntington Disease/microbiology , Myelin Sheath/pathology , Neuronal Plasticity/physiology , White Matter/pathology , Animals , Disease Models, Animal , Huntington Disease/pathology , Mice
5.
Trends Mol Med ; 29(10): 802-816, 2023 10.
Article in English | MEDLINE | ID: mdl-37591764

ABSTRACT

Oligodendrocytes (OLGs), highly specialized glial cells that wrap axons with myelin sheaths, are critical for brain development and function. There is new recognition of the role of OLGs in the pathogenesis of neurodegenerative diseases (NDDs), including Huntington's disease (HD), a prototypic NDD caused by a polyglutamine tract expansion in huntingtin (HTT), which results in gain- and loss-of-function effects. Clinically, HD is characterized by a constellation of motor, cognitive, and psychiatric disturbances. White matter (WM) structures, representing myelin-rich regions of the brain, are profoundly affected in HD, and recent findings reveal oligodendroglia dysfunction as an early pathological event. Here, we focus on mechanisms that underlie oligodendroglial deficits and dysmyelination in the progression of the disease, highlighting the pathogenic contributions of mutant HTT (mHTT). We also discuss potential therapeutic implications involving these molecular pathways.


Subject(s)
Huntington Disease , Humans , Huntington Disease/genetics , Huntington Disease/therapy , Oligodendroglia , Myelin Sheath , Neuroglia , Axons
6.
Mol Neurobiol ; 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38079108

ABSTRACT

Environmental deprivation can have deleterious effects on adaptive myelination and oligodendroglia function. Early stage Huntington disease (HD) is characterised by white-matter myelin abnormalities in both humans and animal models. However, whether deprived environments exacerbate myelin-related pathological features of HD is not clearly understood. Here, we investigated the impact of deprivation and social isolation on ultrastructural features of myelin in the corpus callosum of the YAC128 mouse model of HD and wildtype (WT) mice using transmission electron microscopy. HD pathology on its own leads to increased representation of altered myelin features, such as thinner sheaths and compromised morphology. Interestingly, deprivation mirrors these effects in WT mice but does not greatly exacerbate the already aberrant myelin in HD mice, indicating a disease-related floor effect in the latter animals. These novel findings indicate that environmental deprivation causes abnormalities in myelin ultrastructure in the otherwise healthy corpus callosum of wild-type mice but has distinct effects on HD mice, where compromised myelin integrity is manifest from early stages of the disease.

7.
Nat Neurosci ; 26(12): 2158-2170, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37919424

ABSTRACT

Neuronal homeostasis prevents hyperactivity and hypoactivity. Age-related hyperactivity suggests homeostasis may be dysregulated in later life. However, plasticity mechanisms preventing age-related hyperactivity and their efficacy in later life are unclear. We identify the adult cortical plasticity response to elevated activity driven by sensory overstimulation, then test how plasticity changes with age. We use in vivo two-photon imaging of calcium-mediated cellular/synaptic activity, electrophysiology and c-Fos-activity tagging to show control of neuronal activity is dysregulated in the visual cortex in late adulthood. Specifically, in young adult cortex, mGluR5-dependent population-wide excitatory synaptic weakening and inhibitory synaptogenesis reduce cortical activity following overstimulation. In later life, these mechanisms are downregulated, so that overstimulation results in synaptic strengthening and elevated activity. We also find overstimulation disrupts cognition in older but not younger animals. We propose that specific plasticity mechanisms fail in later life dysregulating neuronal microcircuit homeostasis and that the age-related response to overstimulation can impact cognitive performance.


Subject(s)
Neurons , Visual Cortex , Animals , Neurons/physiology , Homeostasis/physiology , Visual Cortex/physiology , Neuronal Plasticity/physiology
8.
Nat Commun ; 14(1): 5247, 2023 08 28.
Article in English | MEDLINE | ID: mdl-37640701

ABSTRACT

Microglial activation plays central roles in neuroinflammatory and neurodegenerative diseases. Positron emission tomography (PET) targeting 18 kDa Translocator Protein (TSPO) is widely used for localising inflammation in vivo, but its quantitative interpretation remains uncertain. We show that TSPO expression increases in activated microglia in mouse brain disease models but does not change in a non-human primate disease model or in common neurodegenerative and neuroinflammatory human diseases. We describe genetic divergence in the TSPO gene promoter, consistent with the hypothesis that the increase in TSPO expression in activated myeloid cells depends on the transcription factor AP1 and is unique to a subset of rodent species within the Muroidea superfamily. Finally, we identify LCP2 and TFEC as potential markers of microglial activation in humans. These data emphasise that TSPO expression in human myeloid cells is related to different phenomena than in mice, and that TSPO-PET signals in humans reflect the density of inflammatory cells rather than activation state.


Subject(s)
Microglia , Neurodegenerative Diseases , Animals , Mice , Neurodegenerative Diseases/genetics , Macrophages , Myeloid Cells , Genetic Drift
9.
Brain Pathol ; 32(5): e13064, 2022 09.
Article in English | MEDLINE | ID: mdl-35285112

ABSTRACT

Ermin is an actin-binding protein found almost exclusively in the central nervous system (CNS) as a component of myelin sheaths. Although Ermin has been predicted to play a role in the formation and stability of myelin sheaths, this has not been directly examined in vivo. Here, we show that Ermin is essential for myelin sheath integrity and normal saltatory conduction. Loss of Ermin in mice caused de-compacted and fragmented myelin sheaths and led to slower conduction along with progressive neurological deficits. RNA sequencing of the corpus callosum, the largest white matter structure in the CNS, pointed to inflammatory activation in aged Ermin-deficient mice, which was corroborated by increased levels of microgliosis and astrogliosis. The inflammatory milieu and myelin abnormalities were further associated with increased susceptibility to immune-mediated demyelination insult in Ermin knockout mice. Supporting a possible role of Ermin deficiency in inflammatory white matter disorders, a rare inactivating mutation in the ERMN gene was identified in multiple sclerosis patients. Our findings demonstrate a critical role for Ermin in maintaining myelin integrity. Given its near-exclusive expression in myelinating oligodendrocytes, Ermin deficiency represents a compelling "inside-out" model of inflammatory dysmyelination and may offer a new paradigm for the development of myelin stability-targeted therapies.


Subject(s)
Demyelinating Diseases , Multiple Sclerosis , Animals , Central Nervous System/metabolism , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Mice , Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism
10.
Curr Biol ; 31(11): R721-R723, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34102120

ABSTRACT

A new study explores the neural-circuit and synaptic processes that support the transition from general to specific aversive memory formation. A critical role for homeostatic synaptic down-scaling in shaping the specificity of an associative memory is identified.


Subject(s)
Learning , Homeostasis
11.
Cell Calcium ; 94: 102358, 2021 03.
Article in English | MEDLINE | ID: mdl-33517250

ABSTRACT

Aging is a complex process that differentially impacts multiple cognitive, sensory, neuronal and molecular processes. Technological innovations now allow for parallel investigation of neuronal circuit function, structure and molecular composition in the brain of awake behaving adult mice. Thus, mice have become a critical tool to better understand how aging impacts the brain. However, a more granular systems-based approach, which considers the impact of age on key features relating to neural processing, is required. Here, we review evidence probing the impact of age on the mouse brain. We focus on a range of processes relating to neuronal function, including cognitive abilities, sensory systems, synaptic plasticity and calcium regulation. Across many systems, we find evidence for prominent age-related dysregulation even before 12 months of age, suggesting that emerging age-related alterations can manifest by late adulthood. However, we also find reports suggesting that some processes are remarkably resilient to aging. The evidence suggests that aging does not drive a parallel, linear dysregulation of all systems, but instead impacts some processes earlier, and more severely, than others. We propose that capturing the more fine-scale emerging features of age-related vulnerability and resilience may provide better opportunities for the rejuvenation of the aged brain.


Subject(s)
Aging/physiology , Brain/physiology , Calcium/metabolism , Cognition/physiology , Nerve Net/physiology , Animals , Mice , Synapses/physiology
12.
Cell Calcium ; 95: 102365, 2021 05.
Article in English | MEDLINE | ID: mdl-33610083

ABSTRACT

The adult neocortex is not hard-wired but instead retains the capacity to reorganise across multiple spatial scales long into adulthood. Plastic reorganisation occurs at the level of mesoscopic sensory maps, functional neuronal assemblies and synaptic ensembles and is thought to be a critical feature of neuronal network function. Here, we describe a series of approaches that use calcium imaging to measure network reorganisation across multiple spatial scales in vivo. At the mesoscopic level, we demonstrate that sensory activity can be measured in animals undergoing longitudinal behavioural assessment involving automated touchscreen tasks. At the cellular level, we show that network dynamics can be longitudinally measured at both stable and transient functional assemblies. At the level of single synapses, we show that functional subcellular calcium imaging approaches can be used to measure synaptic ensembles of dendritic spines in vivo. Finally, we demonstrate that all three levels of imaging can be spatially related to local pathology in a preclinical rodent model of amyloidosis. We propose that multi-scale in vivo calcium imaging can be used to measure parallel plasticity processes operating across multiple spatial scales in both the healthy brain and preclinical models of disease.


Subject(s)
Amyloidosis/metabolism , Calcium/metabolism , Microscopy, Fluorescence, Multiphoton/methods , Neocortex/metabolism , Nerve Net/metabolism , Amyloidosis/diagnostic imaging , Animals , Disease Models, Animal , Female , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/diagnostic imaging , Nerve Net/diagnostic imaging
13.
Cell Death Dis ; 11(9): 809, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32978366

ABSTRACT

Huntington disease (HD) is a hereditary neurodegenerative disorder caused by mutant huntingtin (mHTT). Phosphorylation at serine-421 (pS421) of mHTT has been shown to be neuroprotective in cellular and rodent models. However, the genetic context of these models differs from that of HD patients. Here we employed human pluripotent stem cells (hiPSCs), which express endogenous full-length mHTT. Using genome editing, we generated isogenic hiPSC lines in which the S421 site in mHTT has been mutated into a phospho-mimetic aspartic acid (S421D) or phospho-resistant alanine (S421A). We observed that S421D, rather than S421A, confers neuroprotection in hiPSC-derived neural cells. Although we observed no effect of S421D on mHTT clearance or axonal transport, two aspects previously reported to be impacted by phosphorylation of mHTT at S421, our analysis revealed modulation of several aspects of mitochondrial form and function. These include mitochondrial surface area, volume, and counts, as well as improved mitochondrial membrane potential and oxidative phosphorylation. Our study validates the protective role of pS421 on mHTT and highlights a facet of the relationship between mHTT and mitochondrial changes in the context of human physiology with potential relevance to the pathogenesis of HD.


Subject(s)
Huntington Disease/genetics , Huntington Disease/metabolism , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Animals , Disease Models, Animal , Humans , Mice , Neuroprotection , Phenotype
14.
Biol Psychiatry ; 88(6): 500-511, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32653109

ABSTRACT

BACKGROUND: Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by epigenetic silencing of FMR1 and loss of FMRP expression. Efforts to understand the molecular underpinnings of the disease have been largely performed in rodent or nonisogenic settings. A detailed examination of the impact of FMRP loss on cellular processes and neuronal properties in the context of isogenic human neurons remains lacking. METHODS: Using CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 to introduce indels in exon 3 of FMR1, we generated an isogenic human pluripotent stem cell model of FXS that shows complete loss of FMRP expression. We generated neuronal cultures and performed genome-wide transcriptome and proteome profiling followed by functional validation of key dysregulated processes. We further analyzed neurodevelopmental and neuronal properties, including neurite length and neuronal activity, using multielectrode arrays and patch clamp electrophysiology. RESULTS: We showed that the transcriptome and proteome profiles of isogenic FMRP-deficient neurons demonstrate perturbations in synaptic transmission, neuron differentiation, cell proliferation and ion transmembrane transporter activity pathways, and autism spectrum disorder-associated gene sets. We uncovered key deficits in FMRP-deficient cells demonstrating abnormal neural rosette formation and neural progenitor cell proliferation. We further showed that FMRP-deficient neurons exhibit a number of additional phenotypic abnormalities, including neurite outgrowth and branching deficits and impaired electrophysiological network activity. These FMRP-deficient related impairments have also been validated in additional FXS patient-derived human-induced pluripotent stem cell neural cells. CONCLUSIONS: Using isogenic human pluripotent stem cells as a model to investigate the pathophysiology of FXS in human neurons, we reveal key neural abnormalities arising from the loss of FMRP.


Subject(s)
Autism Spectrum Disorder , Fragile X Syndrome , Induced Pluripotent Stem Cells , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Humans , Neurons
15.
Nat Commun ; 10(1): 5684, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31831751

ABSTRACT

We experience the world through multiple senses simultaneously. To better understand mechanisms of multisensory processing we ask whether inputs from two senses (auditory and visual) can interact and drive plasticity in neural-circuits of the primary visual cortex (V1). Using genetically-encoded voltage and calcium indicators, we find coincident audio-visual experience modifies both the supra and subthreshold response properties of neurons in L2/3 of mouse V1. Specifically, we find that after audio-visual pairing, a subset of multimodal neurons develops enhanced auditory responses to the paired auditory stimulus. This cross-modal plasticity persists over days and is reflected in the strengthening of small functional networks of L2/3 neurons. We find V1 processes coincident auditory and visual events by strengthening functional associations between feature specific assemblies of multimodal neurons during bouts of sensory driven co-activity, leaving a trace of multisensory experience in the cortical network.


Subject(s)
Auditory Cortex/physiology , Auditory Perception/physiology , Nerve Net/physiology , Visual Cortex/physiology , Visual Perception/physiology , Acoustic Stimulation , Animals , Mice , Models, Animal , Models, Biological , Neuronal Plasticity , Neurons/physiology , Photic Stimulation , Sensory Deprivation/physiology
16.
Mol Neurobiol ; 56(6): 4464-4478, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30334188

ABSTRACT

Laquinimod, an immunomodulatory agent under clinical development for Huntington disease (HD), has recently been shown to confer behavioural improvements that are coupled with prevention of atrophy of the white matter (WM)-rich corpus callosum (CC) in the YAC128 HD mice. However, the nature of the WM improvements is not known yet. Here we investigated the effects of laquinimod on HD-related myelination deficits at the cellular, molecular and ultrastructural levels. We showed that laquinimod treatment improves motor learning and motor function deficits in YAC128 HD mice, and confirmed its antidepressant effect even at the lowest dose used. In addition, we demonstrated for the first time the beneficial effects of laquinimod on myelination in the posterior region of the CC where it reversed changes in myelin sheath thickness and rescued Mbp mRNA and protein deficits. Furthermore, the effect of laquinimod on myelin-related gene expression was not region-specific since the levels of the Mbp and Plp1 transcripts were also increased in the striatum. Also, we did not detect changes in immune cell densities or levels of inflammatory genes in 3-month-old YAC128 HD mice, and these were not altered with laquinimod treatment. Thus, the beneficial effects of laquinimod on HD-related myelination abnormalities in YAC128 HD mice do not appear to be dependent on its immunomodulatory activity. Altogether, our findings describe the beneficial effects of laquinimod treatment on HD-related myelination abnormalities and highlight its therapeutic potential for the treatment of WM pathology in HD patients.


Subject(s)
Huntington Disease/drug therapy , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Quinolones/therapeutic use , Transcription, Genetic , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Behavior, Animal , Cell Count , Corpus Callosum/drug effects , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Cytochrome P-450 CYP1A1/metabolism , Depression/complications , Depression/drug therapy , Depression/physiopathology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Humans , Huntington Disease/complications , Huntington Disease/genetics , Huntington Disease/physiopathology , Inflammation/genetics , Inflammation/pathology , Learning , Male , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Activity/drug effects , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phenotype , Quinolones/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Transcription, Genetic/drug effects
17.
Bio Protoc ; 7(22): e2609, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-34595282

ABSTRACT

Neuronal electrical properties are often aberrant in neurological disorders. Human induced pluripotent stem cells (hiPSCs)-derived neurons represent a useful platform for neurological disease modeling, drug discovery and toxicity screening in vitro. Multi-electrode array (MEA) systems offer a non-invasive and label-free platform to record neuronal evoked-responses concurrently from multiple electrodes. To better detect the neural network changes, we used the Axion Maestro MEA platform to assess neuronal activity and bursting behaviors in hiPSC-derived neuronal cultures. Here we describe the detailed protocol for neuronal culture preparation, MEA recording, and data analysis, which we hope will benefit other researchers in the field.

SELECTION OF CITATIONS
SEARCH DETAIL