Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nat Immunol ; 25(3): 396-398, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38307995

Subject(s)
Ischemic Stroke , Humans , Brain
2.
Immunity ; 55(12): 2318-2335.e7, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36379210

ABSTRACT

Microglia utilize their phagocytic activity to prune redundant synapses and refine neural circuits during precise developmental periods. However, the neuronal signals that control this phagocytic clockwork remain largely undefined. Here, we show that neuronal signal-regulatory protein alpha (SIRPα) is a permissive cue for microglial phagocytosis in the developing murine retina. Removal of neuronal, but not microglial, SIRPα reduced microglial phagocytosis, increased synpase numbers, and impaired circuit function. Conversely, prolonging neuronal SIRPα expression extended developmental microglial phagocytosis. These outcomes depended on the interaction of presynaptic SIRPα with postsynaptic CD47. Global CD47 deficiency modestly increased microglial phagocytosis, while CD47 overexpression reduced it. This effect was rescued by coexpression of neuronal SIRPα or codeletion of neuronal SIRPα and CD47. These data indicate that neuronal SIRPα regulates microglial phagocytosis by limiting microglial SIRPα access to neuronal CD47. This discovery may aid our understanding of synapse loss in neurological diseases.


Subject(s)
CD47 Antigen , Receptors, Immunologic , Mice , Animals , CD47 Antigen/metabolism , Receptors, Immunologic/metabolism , Macrophages/metabolism , Phagocytosis/physiology , Retina , Antigens, Differentiation/metabolism
3.
EMBO Rep ; 24(12): e57164, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37965920

ABSTRACT

A high-salt diet (HSD) elicits sustained sterile inflammation and worsens tissue injury. However, how this occurs after stroke, a leading cause of morbidity and mortality, remains unknown. Here, we report that HSD impairs long-term brain recovery after intracerebral hemorrhage, a severe form of stroke, despite salt withdrawal prior to the injury. Mechanistically, HSD induces innate immune priming and training in hematopoietic stem and progenitor cells (HSPCs) by downregulation of NR4a family and mitochondrial oxidative phosphorylation. This training compromises alternative activation of monocyte-derived macrophages (MDMs) without altering the initial inflammatory responses of the stroke brain. Healthy mice transplanted with bone marrow from HSD-fed mice retain signatures of reduced MDM reparative functions, further confirming a persistent form of innate immune memory that originates in the bone marrow. Loss of NR4a1 in macrophages recapitulates HSD-induced negative impacts on stroke outcomes while gain of NR4a1 enables stroke recovery in HSD animals. Together, we provide the first evidence that links HSD-induced innate immune memory to the acquisition of persistent dysregulated inflammatory responses and unveils NR4a1 as a potential therapeutic target.


Subject(s)
Stroke , Trained Immunity , Mice , Animals , Macrophages , Inflammation , Sodium Chloride, Dietary/adverse effects , Diet , Immunity, Innate
4.
Dev Biol ; 476: 218-239, 2021 08.
Article in English | MEDLINE | ID: mdl-33848537

ABSTRACT

Synapses in the outer retina are the first information relay points in vision. Here, photoreceptors form synapses onto two types of interneurons, bipolar cells and horizontal cells. Because outer retina synapses are particularly large and highly ordered, they have been a useful system for the discovery of mechanisms underlying synapse specificity and maintenance. Understanding these processes is critical to efforts aimed at restoring visual function through repairing or replacing neurons and promoting their connectivity. We review outer retina neuron synapse architecture, neural migration modes, and the cellular and molecular pathways that play key roles in the development and maintenance of these connections. We further discuss how these mechanisms may impact connectivity in the retina.


Subject(s)
Photoreceptor Cells/cytology , Synapses/metabolism , Vision, Ocular/physiology , Animals , Humans , Interneurons/physiology , Photoreceptor Cells/physiology , Retina/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Horizontal Cells/physiology , Synapses/physiology
5.
PLoS Biol ; 15(7): e2001246, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28683067

ABSTRACT

Clinical and preclinical studies indicate that early postnatal exposure to anesthetics can lead to lasting deficits in learning and other cognitive processes. The mechanism underlying this phenomenon has not been clarified and there is no treatment currently available. Recent evidence suggests that anesthetics might cause persistent deficits in cognitive function by disrupting key events in brain development. The hippocampus, a brain region that is critical for learning and memory, contains a large number of neurons that develop in the early postnatal period, which are thus vulnerable to perturbation by anesthetic exposure. Using an in vivo mouse model we demonstrate abnormal development of dendrite arbors and dendritic spines in newly generated dentate gyrus granule cell neurons of the hippocampus after a clinically relevant isoflurane anesthesia exposure conducted at an early postnatal age. Furthermore, we find that isoflurane causes a sustained increase in activity in the mechanistic target of rapamycin pathway, and that inhibition of this pathway with rapamycin not only reverses the observed changes in neuronal development, but also substantially improves performance on behavioral tasks of spatial learning and memory that are impaired by isoflurane exposure. We conclude that isoflurane disrupts the development of hippocampal neurons generated in the early postnatal period by activating a well-defined neurodevelopmental disease pathway and that this phenotype can be reversed by pharmacologic inhibition.


Subject(s)
Anesthetics, Inhalation/toxicity , Cognitive Dysfunction/chemically induced , Hippocampus/drug effects , Isoflurane/toxicity , TOR Serine-Threonine Kinases/metabolism , Animals , Dendritic Spines/drug effects , Dendritic Spines/pathology , Environmental Exposure , Hippocampus/growth & development , Hippocampus/pathology , Mice , Neurons/drug effects , Neurons/pathology
6.
Cell Rep Methods ; 2(7): 100253, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35880013

ABSTRACT

Fine-scale molecular architecture is critical for nervous system and other biological functions. Methods to visualize these nanoscale structures would benefit from enhanced accessibility, throughput, and tissue compatibility. Here, we report RAIN-STORM, a rapid and scalable nanoscopic imaging optimization approach that improves three-dimensional visualization for subcellular targets in tissue at depth. RAIN-STORM uses conventional tissue samples and readily available reagents and is suitable for commercial instrumentation. To illustrate the efficacy of RAIN-STORM, we utilized the retina. We show that RAIN-STORM imaging is versatile and provide 3D nanoscopic data for over 20 synapse, neuron, glia, and vasculature targets. Sample preparation is also rapid, with a 1-day turnaround from tissue to image, and parameters are suitable for multiple tissue sources. Finally, we show that this method can be applied to clinical samples to reveal nanoscale features of human cells and synapses. RAIN-STORM thus paves the way for high-throughput studies of nanoscopic targets in tissue.


Subject(s)
Imaging, Three-Dimensional , Neurons , Humans , Microscopy, Fluorescence , Neuroglia , Synapses
7.
Dis Model Mech ; 15(9)2022 09 01.
Article in English | MEDLINE | ID: mdl-35972048

ABSTRACT

Mutations in the potassium channel tetramerization domain-containing 7 (KCTD7) gene are associated with a severe neurodegenerative phenotype characterized by childhood onset of progressive and intractable myoclonic seizures accompanied by developmental regression. KCTD7-driven disease is part of a large family of progressive myoclonic epilepsy syndromes displaying a broad spectrum of clinical severity. Animal models of KCTD7-related disease are lacking, and little is known regarding how KCTD7 protein defects lead to epilepsy and cognitive dysfunction. We characterized Kctd7 expression patterns in the mouse brain during development and show that it is selectively enriched in specific regions as the brain matures. We further demonstrate that Kctd7-deficient mice develop seizures and locomotor defects with features similar to those observed in human KCTD7-associated diseases. We also show that Kctd7 is required for Purkinje cell survival in the cerebellum and that selective degeneration of these neurons is accompanied by defects in cerebellar microvascular organization and patterning. Taken together, these results define a new model for KCTD7-associated epilepsy and identify Kctd7 as a modulator of neuron survival and excitability linked to microvascular alterations in vulnerable regions.


Subject(s)
Myoclonic Epilepsies, Progressive , Purkinje Cells , Animals , Child , Humans , Mice , Myoclonic Epilepsies, Progressive/genetics , Phenotype , Potassium Channels/genetics , Seizures/genetics
8.
Cell Rep ; 34(5): 108698, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33535040

ABSTRACT

Cone photoreceptors detect light and are responsible for color vision. These cells display a distinct polarized morphology where nuclei are precisely aligned in the apical retina. However, little is known about the mechanisms involved in cone nuclear positioning or the impact of this organization on retina function. We show that the serine/threonine kinase LKB1 and one of its substrates, AMPK, regulate cone nuclear positioning. In the absence of either molecule, cone nuclei are misplaced along the axon, resulting in altered nuclear lamination. LKB1 is required specifically in cones to mediate this process, and disruptions in nuclear alignment result in reduced cone function. Together, these results identify molecular determinants of cone nuclear position and indicate that cone nuclear position alignment enables proper visual function.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Vision, Ocular/physiology , Animals , Mice
9.
Front Neural Circuits ; 14: 583391, 2020.
Article in English | MEDLINE | ID: mdl-33177995

ABSTRACT

During development, neurons generate excess processes which are then eliminated in concert with circuit maturation. C1q is the initiating protein in the complement cascade and has been implicated in this process, but whether C1q-mediated elimination is targeted to particular neural compartments is unclear. Using the murine retina, we identify C1q as a specific regulator of horizontal cell neurite confinement. Subsets of horizontal cell dendritic and axonal neurites extend into the outer retina suggesting that complement achieves both cellular and subcellular selectivity. These alterations emerge as outer retina synapses become mature. C1q expression is restricted to retina microglia, and the loss of C1q results in decreased microglia activation. This pathway appears independent of the C3a receptor (C3aR) and complement receptor 3 (CR3), as horizontal cells are normal when either protein is absent. Together, these data identify a new role for C1q in cell and neurite-specific confinement and implicate microglia-mediated phagocytosis in this process.


Subject(s)
Complement C1q/physiology , Microglia/metabolism , Neurites/physiology , Neuronal Plasticity/physiology , Animals , Complement C3a , Mice , Mice, Knockout , Microglia/physiology , Phagocytosis , Receptors, Complement , Retinal Horizontal Cells
10.
J Comp Neurol ; 528(5): 729-755, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31609468

ABSTRACT

In several areas of the central nervous system, neurons are regionally organized into groups or layers that carry out specific activities. In this form of patterning, neurons of distinct types localize their cell bodies to just one or a few of the layers within a structure. However, little is known about whether diverse neuron types within a lamina share molecular features that coordinate their organization. To begin to identify such candidates, we used the laminated murine retina to screen 92 lacZ reporter lines available through the Knockout Mouse Project. Thirty-two of these displayed reporter expression in restricted subsets of inner retina neurons. We then identified the spatiotemporal expression patterns of these genes at key developmental stages. This uncovered several that were heavily enriched in development but reduced in adulthood, including the transcriptional regulator Hmga1. An additional set of genes displayed maturation associated laminar enrichment. Among these, we identified Bbox1 as a novel gene that specifically labels all neurons in the ganglion cell layer but is largely excluded from otherwise molecularly similar neurons in the inner retina. Finally, we established Dbn1 as a new marker enriched in amacrines and Fmnl3 as a marker for subsets of αRGCs. Together, these data provide a spatiotemporal map for laminae-specific molecules and suggest that diverse neuron types within a lamina share coordinating molecular features that may inform their fate or function.


Subject(s)
Retinal Neurons/cytology , Animals , Mice
11.
Elife ; 92020 05 07.
Article in English | MEDLINE | ID: mdl-32378514

ABSTRACT

Structural changes in pre and postsynaptic neurons that accompany synapse formation often temporally and spatially overlap. Thus, it has been difficult to resolve which processes drive patterned connectivity. To overcome this, we use the laminated outer murine retina. We identify the serine/threonine kinase LKB1 as a key driver of synapse layer emergence. The absence of LKB1 in the retina caused a marked mislocalization and delay in synapse layer formation. In parallel, LKB1 modulated postsynaptic horizontal cell refinement and presynaptic photoreceptor axon growth. Mislocalized horizontal cell processes contacted aberrant cone axons in LKB1 mutants. These defects coincided with altered synapse protein organization, and horizontal cell neurites were misdirected to ectopic synapse protein regions. Together, these data suggest that LKB1 instructs the timing and location of connectivity in the outer retina via coordinate regulation of pre and postsynaptic neuron structure and the localization of synapse-associated proteins.


Subject(s)
Neurites/enzymology , Neurogenesis , Photoreceptor Cells/enzymology , Protein Serine-Threonine Kinases/metabolism , Synapses/enzymology , AMP-Activated Protein Kinases , Animals , Female , Male , Mice, Knockout , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Transport , Vesicular Glutamate Transport Protein 1/metabolism
12.
Neural Dev ; 14(1): 12, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31888774

ABSTRACT

Microglia are increasingly shown to be key players in neuron development and synapse connectivity. However, the underlying mechanisms by which microglia regulate neuron function remain poorly understood in part because such analysis is challenging in the brain where neurons and synapses are intermingled and connectivity is only beginning to be mapped. Here, we discuss the features and function of microglia in the ordered mammalian retina where the laminar organization of neurons and synapses facilitates such molecular studies. We discuss microglia origins and consider the evidence for molecularly distinct microglia subpopulations and their potential for differential roles with a particular focus on the early stages of retina development. We then review the models and methods used for the study of these cells and discuss emerging data that link retina microglia to the genesis and survival of particular retina cell subtypes. We also highlight potential roles for microglia in shaping the development and organization of the vasculature and discuss cellular and molecular mechanisms involved in this process. Such insights may help resolve the mechanisms by which retinal microglia impact visual function and help guide studies of related features in brain development and disease.


Subject(s)
Microglia/physiology , Retina/growth & development , Animals
13.
Neurochem Int ; 129: 104486, 2019 10.
Article in English | MEDLINE | ID: mdl-31175897

ABSTRACT

Neuron function relies on and instructs the development and precise organization of neurovascular units that in turn support circuit activity. However, our understanding of the molecular cues that regulate this relationship remains sparse. Using a high-throughput screening pipeline, we recently identified several new regulators of vascular patterning. Among these was the potassium channel tetramerization domain-containing protein 7 (KCTD7). Mutations in KCTD7 are associated with progressive myoclonic epilepsy, but how KCTD7 regulates neural development and function remains poorly understood. To begin to identify such mechanisms, we focus on mouse retina, a tractable part of the central nervous system that contains precisely ordered neuron subtypes supported by a trilaminar vascular network. We find that deletion of Kctd7 induces defective patterning of the adult retina vascular network, resulting in increased branching, vessel length, and lacunarity. These alterations reflect early and specific defects in vessel development, as emergence of the superficial and deep vascular layers were delayed. These defects are likely due to a role for Kctd7 in inner retina neurons. Kctd7 is absent from vessels but present in neurons in the inner retina, and its deletion resulted in a corresponding increase in the number of bipolar cells in development and increased vessel branching in adults. These alterations were accompanied by retinal function deficits. Together, these data suggest that neuronal Kctd7 drives growth and patterning of the vasculature and that neurovascular interactions may participate in the pathogenesis of KCTD7-related human diseases.


Subject(s)
Potassium Channels/physiology , Retinal Vessels/physiology , Animals , Electroretinography , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoclonic Epilepsies, Progressive/genetics , Potassium Channels/deficiency , Potassium Channels/genetics , RNA, Messenger/biosynthesis , Retina/ultrastructure , Retinal Bipolar Cells/pathology , Retinal Vessels/growth & development , Retinal Vessels/pathology
14.
Cell Rep ; 24(9): 2506-2519, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30157441

ABSTRACT

Retinal function relies on precisely organized neurons and synapses and a properly patterned vasculature to support them. Alterations in these features can result in vision loss. However, our understanding of retinal organization pathways remains incomplete because of a lack of methods to rapidly identify neuron and vasculature regulators in mammals. Here we developed a pipeline for the identification of neural and synaptic integrity genes by high-throughput retinal screening (INSiGHT) that analyzes candidate expression, vascular patterning, cellular organization, and synaptic arrangement. Using this system, we examined 102 mutant mouse lines and identified 16 unique retinal regulatory genes. Fifteen of these candidates are identified as novel retina regulators, and many (9 of 16) are associated with human neural diseases. These results expand the genetic landscape involved in retinal circuit organization and provide a road map for continued discovery of mammalian retinal regulators and disease-causing alleles.


Subject(s)
Neurons/physiology , Retina/physiology , Humans , Synapses
15.
J Neurosurg Anesthesiol ; 28(4): 361-372, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27564556

ABSTRACT

Data from epidemiologic studies and animal models have raised a concern that exposure to anesthetic agents during early postnatal life may cause lasting impairments in cognitive function. It is hypothesized that this is due to disruptions in brain development, but the mechanism underlying this toxic effect remains unknown. Ongoing research, particularly in rodents, has begun to address this question. In this review we examine currently postulated molecular mechanisms of anesthetic toxicity in the developing brain, including effects on cell death pathways, growth factor signaling systems, NMDA and GABA receptors, mitochondria, and epigenetic factors. The level of evidence for each putative mechanism is critically evaluated, and we attempt to draw connections between them where it is possible to do so. Although there are many promising avenues of research, at this time no consensus can be reached as to a definitive mechanism of injury.


Subject(s)
Anesthetics/adverse effects , Brain/drug effects , Neurotoxicity Syndromes/etiology , Animals , Brain/physiopathology , Cell Death/drug effects , Humans , Neurotoxicity Syndromes/physiopathology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL