Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
2.
mBio ; 13(6): e0283822, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36445695

ABSTRACT

Despite recent advances in our understanding of pathogenic access to the central nervous system (CNS), the mechanisms by which intracellular pathogens disseminate within the dense cellular network of neural tissue remain poorly understood. To address this issue, longitudinal analysis of Toxoplasma gondii dissemination in the brain was conducted using 2-photon imaging through a cranial window in living mice that transgenically express enhanced green fluorescent protein (eGFP)-claudin-5. Extracellular T. gondii parasites were observed migrating slowly (1.37 ± 1.28 µm/min) and with low displacement within the brain. In contrast, a population of highly motile infected cells transported vacuoles of T. gondii significantly faster (6.30 ± 3.09 µm/min) and with a higher displacement than free parasites. Detailed analysis of microglial dynamics using CX3CR1-GFP mice revealed that T. gondii-infected microglia remained stationary, and infection did not increase the extension/retraction of microglial processes. The role of infiltrating immune cells in shuttling T. gondii was examined by labeling of peripheral hematopoietic cells with anti-CD45 antibody. Infected CD45+ cells were found crawling along the CNS vessel walls and trafficked T. gondii within the brain parenchyma at significantly higher speeds (3.35 ± 1.70 µm/min) than extracellular tachyzoites. Collectively, these findings highlight a dual role for immune cells in neuroprotection and in facilitating parasite dissemination within the brain. IMPORTANCE T. gondii is a foodborne parasite that infects the brain and can cause fatal encephalitis in immunocompromised individuals. However, there is a limited understanding of how the parasites disseminate through the brain and evade immune clearance. We utilized intravital imaging to visualize extracellular T. gondii tachyzoites and infected cells migrating within the infected mouse brain during acute infection. The infection of motile immune cells infiltrating the brain from the periphery significantly increased the dissemination of T. gondii in the brain compared to that of free parasites migrating using their own motility: the speed and displacement of these infected cells would enable them to cover nearly 1 cm of distance per day! Among the infiltrating cells, T. gondii predominantly infected monocytes and CD8+ T cells, indicating that the parasite can hijack immune cells that are critical for controlling the infection in order to enhance their dissemination within the brain.


Subject(s)
Toxoplasma , Mice , Animals , Toxoplasma/physiology , CD8-Positive T-Lymphocytes , Brain/pathology , Central Nervous System , Monocytes
3.
Curr Biol ; 32(20): 4538-4546.e5, 2022 10 24.
Article in English | MEDLINE | ID: mdl-36152631

ABSTRACT

Despite the recent emergence of multiple cellular and molecular strategies to restore vision in retinal disorders, it remains unclear to what extent central visual circuits can recover when retinal defects are corrected in adulthood. We addressed this question in an Lrat-/- mouse model of Leber congenital amaurosis (LCA) in which retinal light sensitivity and optomotor responses are partially restored by 9-cis-retinyl acetate administration in adulthood. Following treatment, two-photon calcium imaging revealed increases in the number and response amplitude of visually responsive neurons in the primary visual cortex (V1). In particular, retinoid treatment enhanced responses from the ipsilateral eye, restoring the normal balance of eye-specific responses in V1. Additionally, the treatment rescued the modulation of cortical responses by arousal. These findings illustrate the significant plasticity of the adult central visual system and underscore the therapeutic potential of retinoid administration for adults with retinal diseases.


Subject(s)
Retinal Degeneration , Mice , Animals , Retinal Degeneration/drug therapy , Retinoids/pharmacology , Retinoids/therapeutic use , cis-trans-Isomerases , Calcium , Retina , Eye Proteins
4.
Sci Rep ; 12(1): 12779, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35896554

ABSTRACT

Microglia contain multiple mechanisms that shape the synaptic landscape during postnatal development. Whether the synaptic changes mediated by microglia reflect the developmental refinement of neuronal responses in sensory cortices, however, remains poorly understood. In postnatal life, the development of increased orientation and spatial frequency selectivity of neuronal responses in primary visual cortex (V1) supports the emergence of high visual acuity. Here, we used the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 to rapidly and durably deplete microglia in mice during the juvenile period in which increased orientation and spatial frequency selectivity emerge. Excitatory and inhibitory tuning properties were measured simultaneously using multi-photon calcium imaging in layer II/III of mouse V1. We found that microglia depletion generally increased evoked activity which, in turn, reduced orientation selectivity. Surprisingly, microglia were not required for the emergence of high spatial frequency tuned responses. In addition, microglia depletion did not perturb cortical binocularity, suggesting normal depth processing. Together, our finding that orientation and high spatial frequency selectivity in V1 are differentially supported by microglia reveal that microglia are required normal sensory processing, albeit selectively.


Subject(s)
Macrophage Colony-Stimulating Factor/metabolism , Microglia/pathology , Receptors, Colony-Stimulating Factor/physiology , Synapses/physiology , Visual Cortex/physiology , Animals , Mice , Microglia/physiology , Neurons/physiology , Photic Stimulation/methods , Synapses/pathology , Visual Cortex/pathology
5.
Exp Neurol ; 350: 113965, 2022 04.
Article in English | MEDLINE | ID: mdl-34973965

ABSTRACT

This paper explores the potential of rAAV2-retro to deliver gene modifying cargoes to the cells of origin of multiple pathways that are interrupted by spinal cord injury (SCI), summarizing data from previous studies and new data from additional experiments. rAAV-retro exhibits uniquely robust and reliable long-distance retrograde transport from pre-terminal axons and synapses back to neuronal bodies. Previous studies have documented that various AAV-based genetic modifications can enable axon regeneration after SCI, but these have targeted the cells of origin of one pathway at a time. In contrast, rAAV-retro can simultaneously transduce large numbers of neurons of origin of multiple spinal pathways with single injections into the spinal cord. Our initial studies use RosatdTomato and double transgenic PTENf/f; RosatdTomato mice in which transfection with rAAV-retro/Cre deletes PTEN and activates tdT expression in the same neurons. Injections of rAAV-retro/Cre into the cervical, thoracic and lumbar spinal cord led to topographically specific retrograde transduction in cortical motoneurons and neurons in subcortical regions that give rise to different spinal pathways. Our results confirm and extend previous studies indicating selective transduction of neurons that terminate at the level of the injection with minimal retrograde transduction of axons in transit to lower levels. We document feasibility of using rAAV-retro expressing shRNA against PTEN along with a GFP reporter (rAAV-retro-shPTEN/GFP) to effectively knock down PTEN in multiple populations of neurons, which can be used in any species. Some limitations and caveats of currently available rAAV-retros are discussed. Together, our results support the potential applications of rAAV-retro for AAV-based gene-modifications for SCI.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Neural Pathways/growth & development , Spinal Cord Injuries/therapy , Animals , Axons , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Regeneration/genetics , Neural Pathways/injuries , PTEN Phosphohydrolase/genetics , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley
6.
Elife ; 102021 08 23.
Article in English | MEDLINE | ID: mdl-34423781

ABSTRACT

Microglia, the brain's resident myeloid cells, play central roles in brain defense, homeostasis, and disease. Using a prolonged colony-stimulating factor 1 receptor inhibitor (CSF1Ri) approach, we report an unprecedented level of microglial depletion and establish a model system that achieves an empty microglial niche in the adult brain. We identify a myeloid cell that migrates from the subventricular zone and associated white matter areas. Following CSF1Ri, these amoeboid cells migrate radially and tangentially in a dynamic wave filling the brain in a distinct pattern, to replace the microglial-depleted brain. These repopulating cells are enriched in disease-associated microglia genes and exhibit similar phenotypic and transcriptional profiles to white-matter-associated microglia. Our findings shed light on the overlapping and distinct functional complexity and diversity of myeloid cells of the CNS and provide new insight into repopulating microglia function and dynamics in the mouse brain.


Subject(s)
Lateral Ventricles/physiology , Microglia/physiology , White Matter/physiology , Animals , Brain , Disease Models, Animal , Homeostasis , Inflammation , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
7.
Nat Commun ; 12(1): 862, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558487

ABSTRACT

The adult brain lacks sensitivity to changes in the sensory environment found in the juvenile brain. The transplantation of embryonic interneurons has been shown to restore juvenile plasticity to the adult host visual cortex. It is unclear whether transplanted interneurons directly mediate the renewed cortical plasticity or whether these cells act indirectly by modifying the host interneuron circuitry. Here we find that the transplant-induced reorganization of mouse host circuits is specifically mediated by Neuregulin (NRG1)/ErbB4 signaling in host parvalbumin (PV) interneurons. Brief visual deprivation reduces the visual activity of host PV interneurons but has negligible effects on the responses of transplanted PV interneurons. Exogenous NRG1 both prevents the deprivation-induced reduction in the visual responses of host PV interneurons and blocks the transplant-induced reorganization of the host circuit. While deletion of ErbB4 receptors from host PV interneurons blocks cortical plasticity in the transplant recipients, deletion of the receptors from the donor PV interneurons does not. Altogether, our results indicate that transplanted embryonic interneurons reactivate cortical plasticity by rejuvenating the function of host PV interneurons.


Subject(s)
Cell Transplantation , Interneurons/physiology , Interneurons/transplantation , Neuronal Plasticity/physiology , Visual Cortex/embryology , Animals , Cell Differentiation , Dominance, Ocular , Female , Male , Mice, Inbred C57BL , Neuregulin-1/metabolism , Parvalbumins/metabolism , Receptor, ErbB-4/metabolism , Sensory Deprivation , Signal Transduction , Synapses/physiology
8.
J Neurosci Methods ; 350: 109044, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33340556

ABSTRACT

BACKGROUND: The regulation of cerebral blood flow is critical for normal brain functioning, and many physiological and pathological conditions can have long-term impacts on cerebral blood flow. However, minimally invasive tools to study chronic changes in animal models are limited. NEW METHOD: We developed a minimally invasive surgical technique (cyanoacrylate skull, CAS) allowing us to image cerebral blood flow longitudinally through the intact mouse skull using laser speckle imaging. RESULTS: With CAS we were able to detect acute changes in cerebral blood flow induced by hypercapnic challenge. We were also able to image cerebral blood flow dynamics with laser speckle imaging for over 100 days. Furthermore, the relative cerebral blood flow remained stable in mice from 30 days to greater than 100 days after the surgery. COMPARISON WITH EXISTING METHODS: Previously, achieving continuous long-term optical access to measure cerebral blood flow in individual vessels in a mouse model involved invasive surgery. In contrast, the CAS technique presented here is relatively non-invasive, as it allows stable optical access through an intact mouse skull. CONCLUSIONS: The CAS technique allows researcher to chronically measure cerebral blood flow dynamics for a significant portion of a mouse's lifespan. This approach may be useful for studying changes in blood flow due to cerebral pathology or for examining the therapeutic effects of modifying cerebral blood flow in mouse models relevant to human disease.


Subject(s)
Cerebrovascular Circulation , Optical Imaging , Animals , Brain/diagnostic imaging , Brain/surgery , Hemodynamics , Mice , Skull/diagnostic imaging , Skull/surgery
9.
J Neurosci ; 41(7): 1470-1488, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33376158

ABSTRACT

The mammalian visual cortex contains multiple retinotopically defined areas that process distinct features of the visual scene. Little is known about what guides the functional differentiation of visual cortical areas during development. Recent studies in mice have revealed that visual input from the two eyes provides spatiotemporally distinct signals to primary visual cortex (V1), such that contralateral eye-dominated V1 neurons respond to higher spatial frequencies than ipsilateral eye-dominated neurons. To test whether binocular visual input drives the differentiation of visual cortical areas, we used two-photon calcium imaging to characterize the effects of juvenile monocular deprivation (MD) on the responses of neurons in V1 and two higher visual areas, LM (lateromedial) and PM (posteromedial). In adult mice of either sex, we find that MD prevents the emergence of distinct spatiotemporal tuning in V1, LM, and PM. We also find that, within each of these areas, MD reorganizes the distinct spatiotemporal tuning properties driven by the two eyes. Moreover, we find a relationship between speed tuning and ocular dominance in all three areas that MD preferentially disrupts in V1, but not in LM or PM. Together, these results reveal that balanced binocular vision during development is essential for driving the functional differentiation of visual cortical areas. The higher visual areas of mouse visual cortex may provide a useful platform for investigating the experience-dependent mechanisms that set up the specialized processing within neocortical areas during postnatal development.SIGNIFICANCE STATEMENT Little is known about the factors guiding the emergence of functionally distinct areas in the brain. Using in vivo Ca2+ imaging, we recorded visually evoked activity from cells in V1 and higher visual areas LM (lateromedial) and PM (posteromedial) of mice. Neurons in these areas normally display distinct spatiotemporal tuning properties. We found that depriving one eye of normal input during development prevents the functional differentiation of visual areas. Deprivation did not disrupt the degree of speed tuning, a property thought to emerge in higher visual areas. Thus, some properties of visual cortical neurons are shaped by binocular experience, while others are resistant. Our study uncovers the fundamental role of binocular experience in the formation of distinct areas in visual cortex.


Subject(s)
Cell Differentiation/physiology , Vision, Binocular/physiology , Visual Cortex/growth & development , Visual Cortex/physiology , Algorithms , Animals , Brain Mapping , Dominance, Ocular/physiology , Female , Male , Mice , Mice, Inbred C57BL , Neocortex/growth & development , Neocortex/physiology , Neuronal Plasticity , Photic Stimulation , Sensory Deprivation , Space Perception/physiology , Vision, Monocular/physiology , Visual Fields
10.
Cereb Cortex ; 31(5): 2322-2344, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33350438

ABSTRACT

Rostro-caudal specificity of corticospinal tract (CST) projections from different areas of the cortex was assessed by retrograde labeling with fluorogold and retrograde transfection following retro-AAV/Cre injection into the spinal cord of tdT reporter mice. Injections at C5 led to retrograde labeling of neurons throughout forelimb area of the sensorimotor cortex and a region in the dorsolateral cortex near the barrel field (S2). Injections at L2 led to retrograde labeling of neurons in the posterior sensorimotor cortex (hindlimb area) but not the dorsolateral cortex. With injections of biotinylated dextran amine (BDA) into the main sensorimotor cortex (forelimb region), labeled axons terminated selectively at cervical levels. With BDA injections into caudal sensorimotor cortex (hindlimb region), labeled axons passed through cervical levels without sending collaterals into the gray matter and then elaborated terminal arbors at thoracic sacral levels. With BDA injections into the dorsolateral cortex near the barrel field, labeled axons terminated at high cervical levels. Axons from medial sensorimotor cortex terminated primarily in intermediate laminae and axons from lateral sensorimotor cortex terminated primarily in laminae III-V of the dorsal horn. One of the descending pathways seen in rats (the ventral CST) was not observed in most mice.


Subject(s)
Motor Cortex/physiology , Neurons/pathology , Pyramidal Tracts/physiology , Spinal Cord/physiology , Animals , Axons/physiology , Hindlimb/pathology , Hindlimb/physiology , Male , Mice, Inbred BALB C , Motor Cortex/pathology , Neurons/physiology , Pyramidal Tracts/pathology , Spinal Cord/pathology
11.
ACS Chem Biol ; 15(12): 3099-3105, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33222436

ABSTRACT

Profiling RNA expression in a cell-specific manner continues to be a grand challenge in biochemical research. Bioorthogonal nucleosides can be utilized to track RNA expression; however, these methods currently have limitations due to background and incorporation of analogs into undesired cells. Herein, we design and demonstrate that uracil phosphoribosyltransferase can be engineered to match 5-vinyluracil for cell-specific metabolic labeling of RNA with exceptional specificity and stringency.


Subject(s)
RNA/metabolism , Mutation , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Substrate Specificity , Uracil/analogs & derivatives , Uracil/metabolism
12.
Curr Biol ; 30(18): 3591-3603.e8, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32822611

ABSTRACT

Subanesthetic ketamine evokes rapid and long-lasting antidepressant effects in human patients. The mechanism for ketamine's effects remains elusive, but ketamine may broadly modulate brain plasticity processes. We show that single-dose ketamine reactivates adult mouse visual cortical plasticity and promotes functional recovery of visual acuity defects from amblyopia. Ketamine specifically induces downregulation of neuregulin-1 (NRG1) expression in parvalbumin-expressing (PV) inhibitory neurons in mouse visual cortex. NRG1 downregulation in PV neurons co-tracks both the fast onset and sustained decreases in synaptic inhibition to excitatory neurons, along with reduced synaptic excitation to PV neurons in vitro and in vivo following a single ketamine treatment. These effects are blocked by exogenous NRG1 as well as PV targeted receptor knockout. Thus, ketamine reactivation of adult visual cortical plasticity is mediated through rapid and sustained cortical disinhibition via downregulation of PV-specific NRG1 signaling. Our findings reveal the neural plasticity-based mechanism for ketamine-mediated functional recovery from adult amblyopia.


Subject(s)
Amblyopia/drug therapy , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Neuregulin-1/metabolism , Neuronal Plasticity/drug effects , Parvalbumins/metabolism , Visual Cortex/drug effects , Amblyopia/metabolism , Amblyopia/pathology , Animals , Female , Male , Mice , Neuregulin-1/genetics , Neurons/drug effects , Neurons/pathology , Synapses/drug effects , Synapses/pathology , Visual Cortex/pathology
13.
J Neurosci ; 40(3): 585-604, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31767678

ABSTRACT

Study of the neural deficits caused by mismatched binocular vision in early childhood has predominantly focused on circuits in the primary visual cortex (V1). Recent evidence has revealed that neurons in mouse dorsolateral geniculate nucleus (dLGN) can undergo rapid ocular dominance plasticity following monocular deprivation (MD). It remains unclear, however, whether the long-lasting deficits attributed to MD during the critical period originate in the thalamus. Using in vivo two-photon Ca2+ imaging of dLGN afferents in superficial layers of V1 in female and male mice, we demonstrate that 14 d MD during the critical period leads to a chronic loss of binocular dLGN inputs while sparing response strength and spatial acuity. Importantly, MD leads to profoundly mismatched visual tuning properties in remaining binocular dLGN afferents. Furthermore, MD impairs binocular modulation, reducing facilitation of responses of both binocular and monocular dLGN inputs during binocular viewing. As predicted by our findings in thalamic inputs, Ca2+ imaging from V1 neurons revealed spared spatial acuity but impaired binocularity in L4 neurons. V1 L2/3 neurons in contrast displayed deficits in both binocularity and spatial acuity. Our data demonstrate that critical-period MD produces long-lasting disruptions in binocular integration beginning in early binocular circuits in dLGN, whereas spatial acuity deficits first arise from circuits further downstream in V1. Our findings indicate that the development of normal binocular vision and spatial acuity depend upon experience-dependent refinement of distinct stages in the mammalian visual system.SIGNIFICANCE STATEMENT Abnormal binocular vision and reduced acuity are hallmarks of amblyopia, a disorder that affects 2%-5% of the population. It is widely thought that the neural deficits underlying amblyopia begin in the circuits of primary visual cortex. Using in vivo two-photon calcium imaging of thalamocortical axons in mice, we show that depriving one eye of input during a critical period in development chronically impairs binocular integration in thalamic inputs to primary visual cortex. In contrast, visual acuity is spared in thalamic inputs. These findings shed new light on the role for developmental mechanisms in the thalamus in establishing binocular vision and may have critical implications for amblyopia.


Subject(s)
Sensory Deprivation/physiology , Thalamus/growth & development , Thalamus/physiology , Vision, Binocular/physiology , Vision, Monocular/physiology , Vision, Ocular/physiology , Amblyopia/physiopathology , Animals , Brain Mapping , Female , Geniculate Bodies/physiology , Male , Mice , Mice, Inbred C57BL , Photic Stimulation , Space Perception , Visual Acuity/physiology , Visual Cortex/physiology
14.
Proc Natl Acad Sci U S A ; 116(49): 24796-24807, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31727842

ABSTRACT

Brain infection by the parasite Toxoplasma gondii in mice is thought to generate vulnerability to predation by mechanisms that remain elusive. Monocytes play a key role in host defense and inflammation and are critical for controlling T. gondii However, the dynamic and regional relationship between brain-infiltrating monocytes and parasites is unknown. We report the mobilization of inflammatory (CCR2+Ly6Chi) and patrolling (CX3CR1+Ly6Clo) monocytes into the blood and brain during T. gondii infection of C57BL/6J and CCR2RFP/+CX3CR1GFP/+ mice. Longitudinal analysis of mice using 2-photon intravital imaging of the brain through cranial windows revealed that CCR2-RFP monocytes were recruited to the blood-brain barrier (BBB) within 2 wk of T. gondii infection, exhibited distinct rolling and crawling behavior, and accumulated within the vessel lumen before entering the parenchyma. Optical clearing of intact T. gondii-infected brains using iDISCO+ and light-sheet microscopy enabled global 3D detection of monocytes. Clusters of T. gondii and individual monocytes across the brain were identified using an automated cell segmentation pipeline, and monocytes were found to be significantly correlated with sites of T. gondii clusters. Computational alignment of brains to the Allen annotated reference atlas [E. S. Lein et al., Nature 445:168-176 (2007)] indicated a consistent pattern of monocyte infiltration during T. gondii infection to the olfactory tubercle, in contrast to LPS treatment of mice, which resulted in a diffuse distribution of monocytes across multiple brain regions. These data provide insights into the dynamics of monocyte recruitment to the BBB and the highly regionalized localization of monocytes in the brain during T. gondii CNS infection.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/diagnostic imaging , Brain/metabolism , Monocytes/metabolism , Toxoplasmosis/diagnostic imaging , Toxoplasmosis/metabolism , Animals , Antigens, Ly/metabolism , Blood-Brain Barrier/diagnostic imaging , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Receptors, CCR2/metabolism
15.
Neuron ; 103(6): 1016-1033.e10, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31375314

ABSTRACT

iPSC-derived microglia offer a powerful tool to study microglial homeostasis and disease-associated inflammatory responses. Yet, microglia are highly sensitive to their environment, exhibiting transcriptomic deficiencies when kept in isolation from the brain. Furthermore, species-specific genetic variations demonstrate that rodent microglia fail to fully recapitulate the human condition. To address this, we developed an approach to study human microglia within a surrogate brain environment. Transplantation of iPSC-derived hematopoietic-progenitors into the postnatal brain of humanized, immune-deficient mice results in context-dependent differentiation into microglia and other CNS macrophages, acquisition of an ex vivo human microglial gene signature, and responsiveness to both acute and chronic insults. Most notably, transplanted microglia exhibit robust transcriptional responses to Aß-plaques that only partially overlap with that of murine microglia, revealing new, human-specific Aß-responsive genes. We therefore have demonstrated that this chimeric model provides a powerful new system to examine the in vivo function of patient-derived and genetically modified microglia.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cell Differentiation , Gene Expression , Microglia/metabolism , Plaque, Amyloid/genetics , Transplantation Chimera , Animals , Brain/cytology , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Hematopoietic Stem Cell Transplantation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Transgenic , Microglia/cytology , Thrombopoietin/genetics
16.
Sci Rep ; 8(1): 7480, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29748633

ABSTRACT

The end of the critical period for primary visual cortex (V1) coincides with the deposition of perineuronal nets (PNN) onto Parvalbumin (PV) inhibitory neurons. Recently, we found that transplantation of embryonic inhibitory neurons into adult V1 reinstates a new critical period. Here we used Wisteria Floribunda Agglutinin (WFA) staining to compare the deposition of PNNs onto neurons during normal development and following transplantation at equivalent cell ages. In accord with previous findings, PV and PNN expression increases from negligible levels at postnatal day 14 (P14) to mature levels by P70. In contrast to P14, PNNs are found on transplanted PV neurons by 21 days after transplantation and persist to 105 days after transplantation. This precocious deposition was specific to PV neurons and excluded transplanted neurons expressing Somatostatin. Notably, the onset of PV expression in transplanted inhibitory neurons follows the timing of PV expression in juvenile V1. Moreover, transplantation has no discernible effect on host PNNs. The precocious deposition of PNNs onto transplanted PV neurons suggests that PNN expression identified by WFA does not reflect neuronal maturity and may be an inaccurate marker for transplant-induced plasticity of cortical circuits.


Subject(s)
Cell Adhesion , Nerve Net/metabolism , Neurons/metabolism , Neurons/transplantation , Parvalbumins/metabolism , Visual Cortex/cytology , Age Factors , Animals , Cell Adhesion/drug effects , Cell Communication/drug effects , Embryo, Mammalian , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/drug effects , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Plant Lectins/metabolism , Plant Lectins/pharmacology , Pregnancy , Receptors, N-Acetylglucosamine/metabolism , Time Factors , Visual Cortex/drug effects
17.
Cell Rep ; 21(8): 2104-2117, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166603

ABSTRACT

Lymphocytes cross vascular boundaries via either disrupted tight junctions (TJs) or caveolae to induce tissue inflammation. In the CNS, Th17 lymphocytes cross the blood-brain barrier (BBB) before Th1 cells; yet this differential crossing is poorly understood. We have used intravital two-photon imaging of the spinal cord in wild-type and caveolae-deficient mice with fluorescently labeled endothelial tight junctions to determine how tight junction remodeling and caveolae regulate CNS entry of lymphocytes during the experimental autoimmune encephalomyelitis (EAE) model for multiple sclerosis. We find that dynamic tight junction remodeling occurs early in EAE but does not depend upon caveolar transport. Moreover, Th1, but not Th17, lymphocytes are significantly reduced in the inflamed CNS of mice lacking caveolae. Therefore, tight junction remodeling facilitates Th17 migration across the BBB, whereas caveolae promote Th1 entry into the CNS. Moreover, therapies that target both tight junction degradation and caveolar transcytosis may limit lymphocyte infiltration during inflammation.


Subject(s)
Blood-Brain Barrier/metabolism , Caveolin 1/metabolism , Inflammation/metabolism , Th1 Cells/immunology , Tight Junctions/metabolism , Animals , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelium, Vascular/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Th17 Cells/immunology
18.
J Neurosci ; 37(42): 10125-10138, 2017 10 18.
Article in English | MEDLINE | ID: mdl-28924011

ABSTRACT

Binocular mechanisms for visual processing are thought to enhance spatial acuity by combining matched input from the two eyes. Studies in the primary visual cortex of carnivores and primates have confirmed that eye-specific neuronal response properties are largely matched. In recent years, the mouse has emerged as a prominent model for binocular visual processing, yet little is known about the spatial frequency tuning of binocular responses in mouse visual cortex. Using calcium imaging in awake mice of both sexes, we show that the spatial frequency preference of cortical responses to the contralateral eye is ∼35% higher than responses to the ipsilateral eye. Furthermore, we find that neurons in binocular visual cortex that respond only to the contralateral eye are tuned to higher spatial frequencies. Binocular neurons that are well matched in spatial frequency preference are also matched in orientation preference. In contrast, we observe that binocularly mismatched cells are more mismatched in orientation tuning. Furthermore, we find that contralateral responses are more direction-selective than ipsilateral responses and are strongly biased to the cardinal directions. The contralateral bias of high spatial frequency tuning was found in both awake and anesthetized recordings. The distinct properties of contralateral cortical responses may reflect the functional segregation of direction-selective, high spatial frequency-preferring neurons in earlier stages of the central visual pathway. Moreover, these results suggest that the development of binocularity and visual acuity may engage distinct circuits in the mouse visual system.SIGNIFICANCE STATEMENT Seeing through two eyes is thought to improve visual acuity by enhancing sensitivity to fine edges. Using calcium imaging of cellular responses in awake mice, we find surprising asymmetries in the spatial processing of eye-specific visual input in binocular primary visual cortex. The contralateral visual pathway is tuned to higher spatial frequencies than the ipsilateral pathway. At the highest spatial frequencies, the contralateral pathway strongly prefers to respond to visual stimuli along the cardinal (horizontal and vertical) axes. These results suggest that monocular, and not binocular, mechanisms set the limit of spatial acuity in mice. Furthermore, they suggest that the development of visual acuity and binocularity in mice involves different circuits.


Subject(s)
Orientation/physiology , Photic Stimulation/methods , Space Perception/physiology , Visual Cortex/physiology , Visual Pathways/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
19.
J Neurosci ; 37(4): 820-829, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28123018

ABSTRACT

The maturation of cortical parvalbumin-positive (PV) interneurons depends on the interaction of innate and experience-dependent factors. Dark-rearing experiments suggest that visual experience determines when broad orientation selectivity emerges in visual cortical PV interneurons. Here, using neural transplantation and in vivo calcium imaging of mouse visual cortex, we investigated whether innate mechanisms contribute to the maturation of orientation selectivity in PV interneurons. First, we confirmed earlier findings showing that broad orientation selectivity emerges in PV interneurons by 2 weeks after vision onset, ∼35 d after these cells are born. Next, we assessed the functional development of transplanted PV (tPV) interneurons. Surprisingly, 25 d after transplantation (DAT) and >2 weeks after vision onset, we found that tPV interneurons have not developed broad orientation selectivity. By 35 DAT, however, broad orientation selectivity emerges in tPV interneurons. Transplantation does not alter orientation selectivity in host interneurons, suggesting that the maturation of tPV interneurons occurs independently from their endogenous counterparts. Together, these results challenge the notion that the onset of vision solely determines when PV interneurons become broadly tuned. Our results reveal that an innate cortical mechanism contributes to the emergence of broad orientation selectivity in PV interneurons. SIGNIFICANCE STATEMENT: Early visual experience and innate developmental programs interact to shape cortical circuits. Visual-deprivation experiments have suggested that the onset of visual experience determines when interneurons mature in the visual cortex. Here we used neuronal transplantation and cellular imaging of visual responses to investigate the maturation of parvalbumin-positive (PV) interneurons. Our results suggest that the emergence of broad orientation selectivity in PV interneurons is innately timed.


Subject(s)
Interneurons/physiology , Orientation/physiology , Parvalbumins/physiology , Photic Stimulation/methods , Visual Cortex/cytology , Visual Cortex/growth & development , Animals , Female , Male , Mice , Mice, Transgenic
20.
Neuron ; 92(6): 1157-1159, 2016 Dec 21.
Article in English | MEDLINE | ID: mdl-28009270

ABSTRACT

Yang et al. (2016) show that transplantation of GABAergic inhibitory neurons into the amygdala boosts the persistence of fear extinction in mice. Transplantation was found to degrade perineuronal nets on endogenous inhibitory neurons and enhance synaptic plasticity in host amygdala.


Subject(s)
Amygdala , Fear , Animals , Mice , Neuronal Plasticity , Neurons
SELECTION OF CITATIONS
SEARCH DETAIL
...