Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.017
Filter
1.
Bull Exp Biol Med ; 176(5): 666-671, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38727956

ABSTRACT

This paper shows for the first time that co-transplantation of human olfactory ensheathing cells with neurotrophin-3 into spinal cord cysts is more effective for activation of remyelination than transplantation of cells with brain-derived neurotrophic factor and a combination of these two factors. The studied neurotrophic factors do not affect proliferation and migration of ensheathing cells in vitro. It can be concluded that the maximum improvement of motor function in rats receiving ensheathing cells with neurotrophin-3 is largely determined by activation of remyelination.


Subject(s)
Brain-Derived Neurotrophic Factor , Neurotrophin 3 , Olfactory Bulb , Remyelination , Animals , Rats , Neurotrophin 3/metabolism , Humans , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Remyelination/physiology , Olfactory Bulb/cytology , Cell Proliferation , Spinal Cord/metabolism , Myelin Sheath/metabolism , Myelin Sheath/physiology , Cells, Cultured , Cell Movement , Cysts/pathology , Female , Central Nervous System Cysts/surgery , Central Nervous System Cysts/pathology
2.
Proc Natl Acad Sci U S A ; 121(20): e2321711121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38713624

ABSTRACT

During development, neural stem cells in the cerebral cortex, also known as radial glial cells (RGCs), generate excitatory neurons, followed by production of cortical macroglia and inhibitory neurons that migrate to the olfactory bulb (OB). Understanding the mechanisms for this lineage switch is fundamental for unraveling how proper numbers of diverse neuronal and glial cell types are controlled. We and others recently showed that Sonic Hedgehog (Shh) signaling promotes the cortical RGC lineage switch to generate cortical oligodendrocytes and OB interneurons. During this process, cortical RGCs generate intermediate progenitor cells that express critical gliogenesis genes Ascl1, Egfr, and Olig2. The increased Ascl1 expression and appearance of Egfr+ and Olig2+ cortical progenitors are concurrent with the switch from excitatory neurogenesis to gliogenesis and OB interneuron neurogenesis in the cortex. While Shh signaling promotes Olig2 expression in the developing spinal cord, the exact mechanism for this transcriptional regulation is not known. Furthermore, the transcriptional regulation of Olig2 and Egfr has not been explored. Here, we show that in cortical progenitor cells, multiple regulatory programs, including Pax6 and Gli3, prevent precocious expression of Olig2, a gene essential for production of cortical oligodendrocytes and astrocytes. We identify multiple enhancers that control Olig2 expression in cortical progenitors and show that the mechanisms for regulating Olig2 expression are conserved between the mouse and human. Our study reveals evolutionarily conserved regulatory logic controlling the lineage switch of cortical neural stem cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cerebral Cortex , ErbB Receptors , Hedgehog Proteins , Nerve Tissue Proteins , Neural Stem Cells , Neurogenesis , Oligodendrocyte Transcription Factor 2 , PAX6 Transcription Factor , Animals , Neurogenesis/physiology , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Mice , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendrocyte Transcription Factor 2/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , PAX6 Transcription Factor/metabolism , PAX6 Transcription Factor/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Eye Proteins/metabolism , Eye Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/genetics , Neuroglia/metabolism , Neuroglia/cytology , Gene Expression Regulation, Developmental , Signal Transduction , Olfactory Bulb/metabolism , Olfactory Bulb/cytology , Cell Lineage , Humans
3.
Auris Nasus Larynx ; 51(3): 517-524, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38522356

ABSTRACT

OBJECTIVE: Periglomerular and granule cells in the adult mammalian olfactory bulb modulate olfactory signal transmission. These cells originate from the subventricular zone, migrate to the olfactory bulb via the Rostral Migratory Stream (RMS), and differentiate into mature cells within the olfactory bulb throughout postnatal life. While the regulation of neuroblast development is known to be affected by external stimuli, there is a lack of information concerning changes that occur during the recovery process after injury caused by external stimuli. To address this gap in research, the present study conducted histological observations to investigate changes in the olfactory bulb and RMS occurring after the degeneration and regeneration of olfactory neurons. METHODS: To create a model of olfactory neurodegeneration, adult mice were administered methimazole intraperitoneally. Nasal tissue and whole brains were removed 3, 7, 14 and 28 days after methimazole administration, and EdU was administered 2 and 4 h before removal of these tissues to monitor dividing cells in the RMS. Methimazole-untreated mice were used as controls. Olfactory nerve fibers entering the olfactory glomerulus were observed immunohistochemically using anti-olfactory marker protein. In the brain tissue, the entire RMS was observed and the volume and total number of cells in the RMS were measured. In addition, the number of neuroblasts and dividing neuroblasts passing through the RMS were measured using anti-doublecortin and anti-EdU antibodies, respectively. Statistical analysis was performed using the Tukey test. RESULTS: Olfactory epithelium degenerated was observed after methimazole administration, and recovered after 28 days. In the olfactory glomeruli, degeneration of OMP fibers began after methimazole administration, and after day 14, OMP fibers were reduced or absent by day 28, and overall OMP positive fibers were less than 20%. Glomerular volume tended to decrease after methimazole administration and did not appear to recover, even 28 days after recovery of the olfactory epithelium. In the RMS, EdU-positive cells decreased on day 3 and began to increase on day 7. However, they did not recover to the same levels as the control methimazole-untreated mice even after 28 days. CONCLUSION: These results suggest that the division and maturation of neuroblasts migrating from the RMS was suppressed by olfactory nerve degeneration or the disruption of olfactory input.


Subject(s)
Cell Movement , Methimazole , Olfactory Bulb , Animals , Olfactory Bulb/pathology , Olfactory Bulb/drug effects , Olfactory Bulb/cytology , Methimazole/pharmacology , Mice , Antithyroid Agents/pharmacology , Olfactory Nerve/pathology , Olfactory Marker Protein/metabolism , Disease Models, Animal , Male
4.
J Chem Neuroanat ; 137: 102413, 2024 04.
Article in English | MEDLINE | ID: mdl-38492895

ABSTRACT

Chronic pain is often accompanied by tissue damage and pain hypersensitivity. It easily relapses and is challenging to cure, which seriously affects the patients' quality of life and is an urgent problem to be solved. Current treatment methods primarily rely on morphine drugs, which do not address the underlying nerve injury and may cause adverse reactions. Therefore, in recent years, scientists have shifted their focus from chronic pain treatment to cell transplantation. This review describes the classification and mechanism of chronic pain through the introduction of the characteristics of olfactory ensheathing cells (OECs), an in-depth discussion of special glial cells through the phagocytosis of nerve debris, receptor-ligand interactions, providing nutrition, and other inhibition of neuroinflammation, and ultimately supporting axon regeneration and mitigation of chronic pain. This review summarizes the potential and limitations of OECs for treating chronic pain by objectively analyzing relevant clinical trials and methods to enhance efficacy and future development prospects.


Subject(s)
Chronic Pain , Olfactory Bulb , Humans , Chronic Pain/therapy , Animals , Olfactory Bulb/cytology , Neuroglia , Cell Transplantation/methods
5.
Science ; 382(6673): 958-963, 2023 11 24.
Article in English | MEDLINE | ID: mdl-37995223

ABSTRACT

Adult neural stem cells (NSCs) contribute to lifelong brain plasticity. In the adult mouse ventricular-subventricular zone, NSCs are heterogeneous and, depending on their location in the niche, give rise to different subtypes of olfactory bulb (OB) interneurons. Here, we show that multiple regionally distinct NSCs, including domains that are usually quiescent, are recruited on different gestation days during pregnancy. Synchronized activation of these adult NSC pools generates transient waves of short-lived OB interneurons, especially in layers with less neurogenesis under homeostasis. Using spatial transcriptomics, we identified molecular markers of pregnancy-associated interneurons and showed that some subsets are temporarily needed for own pup recognition. Thus, pregnancy triggers transient yet behaviorally relevant neurogenesis, highlighting the physiological relevance of adult stem cell heterogeneity.


Subject(s)
Interneurons , Lateral Ventricles , Maternal Behavior , Neurogenesis , Neuronal Plasticity , Olfactory Bulb , Pregnancy , Smell , Animals , Female , Mice , Pregnancy/physiology , Adult Stem Cells/physiology , Interneurons/cytology , Interneurons/physiology , Lateral Ventricles/cytology , Lateral Ventricles/growth & development , Neural Stem Cells/physiology , Neurogenesis/physiology , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , Olfactory Bulb/metabolism , Transcriptome , Maternal Behavior/physiology
6.
Sci Rep ; 12(1): 1875, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115567

ABSTRACT

Genetically encoded voltage indicators (GEVIs) allow optical recordings of membrane potential changes in defined cell populations. Transgenic reporter animals that facilitate precise and repeatable targeting with high expression levels would further the use of GEVIs in the in vivo mammalian brain. However, the literature on developing and applying transgenic mouse lines as vehicles for GEVI expression is limited. Here we report the first in vivo experiments using a transgenic reporter mouse for the GEVI ArcLight, which utilizes a Cre/tTA dependent expression system (TIGRE 1.0). We developed two mouse lines with ArcLight expression restricted to either olfactory receptor neurons, or a subpopulation of interneurons located in the granule and glomerular layers in the olfactory bulb. The ArcLight expression in these lines was sufficient for in vivo imaging of odorant responses in single trials using epifluorescence and 2-photon imaging. The voltage responses were odor-specific and concentration-dependent, which supported earlier studies about perceptual transformations carried out by the bulb that used calcium sensors of neural activity. This study demonstrates that the ArcLight transgenic line is a flexible genetic tool that can be used to record the neuronal electrical activity of different cell types with a signal-to-noise ratio that is comparable to previous reports using viral transduction.


Subject(s)
Biosensing Techniques , Interneurons/metabolism , Luminescent Proteins/metabolism , Membrane Potentials , Olfactory Bulb/metabolism , Olfactory Receptor Neurons/metabolism , Recombinant Fusion Proteins/metabolism , Voltage-Sensitive Dye Imaging , Animals , Genes, Reporter , Luminescent Proteins/genetics , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Odorants , Olfactory Bulb/cytology , Olfactory Perception , Recombinant Fusion Proteins/genetics , Smell
7.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-35023540

ABSTRACT

Estrogens are well-known to regulate development of sexual dimorphism of the brain; however, their role in embryonic brain development prior to sex-differentiation is unclear. Using estrogen biosensor zebrafish models, we found that estrogen activity in the embryonic brain occurs from early neurogenesis specifically in a type of glia in the olfactory bulb (OB), which we name estrogen-responsive olfactory bulb (EROB) cells. In response to estrogen, EROB cells overlay the outermost layer of the OB and interact tightly with olfactory sensory neurons at the olfactory glomeruli. Inhibiting estrogen activity using an estrogen receptor antagonist, ICI182,780 (ICI), and/or EROB cell ablation impedes olfactory glomerular development, including the topological organisation of olfactory glomeruli and inhibitory synaptogenesis in the OB. Furthermore, activation of estrogen signalling inhibits both intrinsic and olfaction-dependent neuronal activity in the OB, whereas ICI or EROB cell ablation results in the opposite effect on neuronal excitability. Altering the estrogen signalling disrupts olfaction-mediated behaviour in later larval stage. We propose that estrogens act on glia to regulate development of OB circuits, thereby modulating the local excitability in the OB and olfaction-mediated behaviour.


Subject(s)
Estrogens/metabolism , Neurogenesis , Neuroglia/cytology , Olfactory Bulb/embryology , Animals , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Neuroglia/drug effects , Neuroglia/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/drug effects , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Receptors, Estrogen/antagonists & inhibitors , Synapses/metabolism , Synapses/physiology , Zebrafish
8.
Mol Neurobiol ; 59(1): 77-92, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34625907

ABSTRACT

Human MYCN is an oncogene amplified in neuroblastoma and many other tumors. Both human MYCN and mouse Mycn genes are important in embryonic brain development, but their functions in adult healthy nerve system are completely unknown. Here, with Mycn-eGFP mice and quantitative RT-PCR, we found that Mycn was expressed in specific brain regions of young adult mice, including subventricular zone (SVZ), subgranular zone (SGZ), olfactory bulb (OB), subcallosal zone (SCZ), and corpus callosum (CC). With immunohistochemistry (IHC), we found that many Mycn-expressing cells expressed neuroblast marker doublecortin (DCX) and proliferation marker Ki67. With Dcx-creER and Mki67-creER mouse lines, we fate mapped Dcx-expressing neuroblasts and Mki67-expressing proliferation cells, along with deleting Mycn from these cells in adult mice. We found that knocking out Mycn from adult neuroblasts or proliferating cells significantly reduced cells in proliferation in SVZ, SGZ, OB, SCZ, and CC. We also demonstrated that the Mycn-deficient neuroblasts in SGZ matured quicker than wild-type neuroblasts, and that Mycn-deficient proliferating cells were more likely to survive in SVZ, SGZ, OB, SCZ, and CC compared to wild type. Thus, our results demonstrate that, in addition to causing tumors in the nervous system, oncogene Mycn has a crucial function in neurogenesis and oligodendrogenesis in adult healthy brain.


Subject(s)
N-Myc Proto-Oncogene Protein/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Oligodendroglia/metabolism , Animals , Cell Proliferation/physiology , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein/genetics , Neural Stem Cells/cytology , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Oligodendroglia/cytology
9.
Brain Res Bull ; 179: 13-24, 2022 02.
Article in English | MEDLINE | ID: mdl-34848271

ABSTRACT

Overweight induced by high-fat diet (HFD) represents one of the major health concerns in modern societies, which can cause lasting peripheral and central metabolic disorders in all age groups. Specifically, childhood obesity could lead to life-long impact on brain development and functioning. On the other hand, environmental enrichment (EE) has been demonstrated to be beneficial for learning and memory. Here, we explored the impact of high-fat diet on olfaction and organization of olfactory bulb cells in adolescent mice, and the effect of EE intervention thereon. Puberty mice (3-week-old) fed with HFD for 10 weeks exhibited poorer odor sensitivity and olfactory memory relative to controls consuming standard chows. The behavioral deficits were rescued in the HFD group with EE intervention. Neuroanatomically, parvalbumin (PV) interneurons in the olfactory bulb (OB) were reduced in the HFD-fed animals relative to control, while EE intervention also normalized this alteration. In contrast, cells expressing calbindin (CB), doublecortin (DCX) in the OB were not altered. Our findings suggest that PV interneurons may play a crucial role in mediating the HFD-induced olfactory deficit in adolescent mice, and can also serve a protective effect of EE against the functional deficit.


Subject(s)
Diet, High-Fat/adverse effects , Environment , Interneurons/metabolism , Olfaction Disorders/etiology , Olfaction Disorders/therapy , Olfactory Bulb , Parvalbumins/metabolism , Age Factors , Animals , Behavior, Animal/physiology , Disease Models, Animal , Mice , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Olfactory Bulb/physiopathology
10.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768936

ABSTRACT

Neural precursors originating in the subventricular zone (SVZ), the largest neurogenic region of the adult brain, migrate several millimeters along a restricted migratory pathway, the rostral migratory stream (RMS), toward the olfactory bulb (OB), where they differentiate into interneurons and integrate into the local neuronal circuits. Migration of SVZ-derived neuroblasts in the adult brain differs in many aspects from that in the embryonic period. Unlike in that period, postnatally-generated neuroblasts in the SVZ are able to divide during migration along the RMS, as well as they migrate independently of radial glia. The homophilic mode of migration, i.e., using each other to move, is typical for neuroblast movement in the RMS. In addition, it has recently been demonstrated that specifically-arranged blood vessels navigate SVZ-derived neuroblasts to the OB and provide signals which promote migration. Here we review the development of vasculature in the presumptive neurogenic region of the rodent brain during the embryonic period as well as the development of the vascular scaffold guiding neuroblast migration in the postnatal period, and the significance of blood vessel reorganization during the early postnatal period for proper migration of RMS neuroblasts in adulthood.


Subject(s)
Brain/blood supply , Lateral Ventricles/physiology , Neovascularization, Physiologic/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Olfactory Bulb/physiology , Animals , Blood Vessels/metabolism , Brain/embryology , Cell Movement/physiology , Lateral Ventricles/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/cytology , Neurons/physiology , Olfactory Bulb/cytology
11.
PLoS Comput Biol ; 17(9): e1009169, 2021 09.
Article in English | MEDLINE | ID: mdl-34543261

ABSTRACT

The majority of olfaction studies focus on orthonasal stimulation where odors enter via the front nasal cavity, while retronasal olfaction, where odors enter the rear of the nasal cavity during feeding, is understudied. The coding of retronasal odors via coordinated spiking of neurons in the olfactory bulb (OB) is largely unknown despite evidence that higher level processing is different than orthonasal. To this end, we use multi-electrode array in vivo recordings of rat OB mitral cells (MC) in response to a food odor with both modes of stimulation, and find significant differences in evoked firing rates and spike count covariances (i.e., noise correlations). Differences in spiking activity often have implications for sensory coding, thus we develop a single-compartment biophysical OB model that is able to reproduce key properties of important OB cell types. Prior experiments in olfactory receptor neurons (ORN) showed retro stimulation yields slower and spatially smaller ORN inputs than with ortho, yet whether this is consequential for OB activity remains unknown. Indeed with these specifications for ORN inputs, our OB model captures the salient trends in our OB data. We also analyze how first and second order ORN input statistics dynamically transfer to MC spiking statistics with a phenomenological linear-nonlinear filter model, and find that retro inputs result in larger linear filters than ortho inputs. Finally, our models show that the temporal profile of ORN is crucial for capturing our data and is thus a distinguishing feature between ortho and retro stimulation, even at the OB. Using data-driven modeling, we detail how ORN inputs result in differences in OB dynamics and MC spiking statistics. These differences may ultimately shape how ortho and retro odors are coded.


Subject(s)
Action Potentials/physiology , Models, Biological , Nasal Cavity/physiology , Olfactory Bulb/physiology , Animals , Odorants , Olfactory Bulb/cytology , Olfactory Receptor Neurons/physiology , Rats
12.
Neurobiol Aging ; 108: 47-57, 2021 12.
Article in English | MEDLINE | ID: mdl-34507271

ABSTRACT

Olfactory damage develops at the early stages of Alzheimer's disease (AD). While amyloid-ß (Aß) oligomers are shown to impair inhibitory circuits in the olfactory bulb (OB), its underlying mechanisms remain unclear. Here, we investigated the olfactory dysfunction due to impaired inhibitory transmission to mitral cells (MCs) of the OB in APP/PS1 mice. Using electrophysiological studies, we found that MCs exhibited increased spontaneous firing rates as early as 3 months, much before development of Aß deposits in the brain. Furthermore, the frequencies but not amplitudes of MC inhibitory postsynaptic currents decreased markedly, suggesting that presynaptic GABA release is impaired while postsynaptic GABAA receptor responses remain intact. Notably, muscimol, a GABAA receptor agonist, improved odor identification and discrimination behaviors in APP/PS1 mice, reduced MC basal firing activity, and rescued inhibitory circuits along with reducing the Aß burden in the OB. Our study links the presynaptic deficits of GABAergic transmission to olfactory dysfunction and subsequent AD development and implicates the therapeutic potential of maintaining local inhibitory microcircuits against early AD progression.


Subject(s)
GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Agonists/therapeutic use , Olfaction Disorders/drug therapy , Olfaction Disorders/physiopathology , Olfactory Bulb/pathology , Olfactory Bulb/physiopathology , Receptors, GABA-A/physiology , Smell/drug effects , Synaptic Transmission/drug effects , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/adverse effects , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Mice, Transgenic , Olfaction Disorders/etiology , Olfactory Bulb/cytology , Presenilin-1/genetics , Time Factors
13.
J Neurochem ; 158(5): 1186-1198, 2021 09.
Article in English | MEDLINE | ID: mdl-34338310

ABSTRACT

During adult rodent life, newborn neurons are added to the olfactory bulb (OB) in a tightly controlled manner. Upon arrival in the OB, input synapses from the local bulbar network and the higher olfactory cortex precede the formation of functional output synapses, indicating a possible role for these regions in newborn neuron survival. An interplay between the environment and the piriform cortex in the regulation of newborn neuron survival has been suggested. However, the specific network and the neuronal cell types responsible for this effect have not been elucidated. Furthermore, the role of the other olfactory cortical areas in this process is not known. Here we demonstrate that pyramidal neurons in the mouse anterior olfactory nucleus, the first cortical area for odor processing, have a key role in the survival of newborn neurons. Using DREADD (Designer Receptors Exclusively Activated by Designer Drugs) technology, we applied chronic stimulation to the anterior olfactory nucleus and observed a decrease in newborn neurons in the OB through induction of apoptosis. These findings provide further insight into the network regulating neuronal survival in adult neurogenesis and strengthen the importance of the surrounding network for sustained integration of new neurons.


Subject(s)
Neurogenesis/physiology , Neurons/physiology , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Olfactory Cortex/cytology , Olfactory Cortex/physiology , Age Factors , Animals , Cell Survival/drug effects , Cell Survival/physiology , Female , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Neurons/drug effects , Odorants , Olfactory Bulb/drug effects , Olfactory Cortex/drug effects , Olfactory Pathways/cytology , Olfactory Pathways/drug effects , Olfactory Pathways/physiology , Smell/physiology
14.
Pharmacol Res ; 172: 105797, 2021 10.
Article in English | MEDLINE | ID: mdl-34352399

ABSTRACT

Since both Olfactory ensheathing cells (OECs) and neural stem cells (NSCs) have shown certain efficacy in the cellular therapy of nerve injury and disease, there have been a series of investigations in recent years looking at the co-culture of NSCs and OECs. Protein phosphorylation forms the basis for identifying a variety of cellular signaling pathways responsible for regulating the self-renewal and differentiation of NSCs induced by OECs. To better understand the signaling cascades in the early phases of OEC-induced NSC differentiation, changes in the NSC proteome and phosphoproteome during the first 24 h were determined using dimethyl labeling and TiO2 phosphorylation enrichment coupled with Liquid chromatography-tandem mass spectrometry (LC-MS/MS). A total of 565 proteins and 2511 phosphorylation sites were identified. According to quantitative phosphoproteomics analyses of NSC differentiation induced by OECs during the first 12 and 24 h, it was speculated that there were at least two different signal waves: one peaking within 12 h after stimulation and the second upsurge after 24 h. In addition to understanding the dynamics of the proteome and phosphoproteome in the early stages of NSC differentiation, our analyses identified a key role of the TGF-ß3 protein secreted by OECs, which may be an initiating factor that promotes differentiation of NSCs into neurons induced by OECs. These findings not only redemonstrated a OECs-based therapeutic strategy in cell therapy, but also added a node to the regulatory network for the neural lineage commitment of NSCs induced by OECs.


Subject(s)
Neural Stem Cells/metabolism , Neuroglia , Olfactory Bulb/cytology , Phosphoproteins/genetics , Proteome/genetics , Animals , Cell Differentiation , Cells, Cultured , Culture Media, Conditioned , Mice , Phosphoproteins/metabolism , Phosphorylation , Protein Interaction Maps , Proteomics
15.
Eur J Pharm Biopharm ; 167: 189-200, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34333085

ABSTRACT

Both dopamine (DA) loaded Solid Lipid Nanoparticles (SLN) and liposomes (Lip), designed for intranasal administration of the neurotransmitter as an innovative Parkinson disease treatment, were already characterized in vitro in some extent by us (Trapani et al., 2018a and Cometa et al., 2020, respectively). Herein, to gain insight into the structure of SLN, X-ray Photoelectron Spectroscopy Analysis was carried out and DA-SLN (SLN 1) were found to exhibit high amounts of the neurotransmitter on the surface, whereas the external side of Glycol Chitosan (GCS) containing SLN (SLN 2) possessed only few amounts. However, SLN 2 were characterized by the highest encapsulation DA efficiency (i.e., 81%). Furthermore, in view of intranasal administration, mucoadhesion tests in vitro were also conducted for SLN and Lip formulations, evidencing high muchoadesive effect exerted by SLN 2. Concerning ex-vivo studies, SLN and Lip were found to be safe for Olfactory Ensheathing Cells and fluorescent SLN 2 were taken up in a dose-dependent manner reaching the 100% of positive cells, while Lip 2 (chitosan-glutathione-coated) were internalised by 70% OECs with six-times more lipid concentration. Hence, SLN 2 formulation containing DA and GCS may constitute interesting formulations for further studies and promising dosage form for non-invasive nose-to-brain neurotransmitter delivery.


Subject(s)
Dopamine Agents/administration & dosage , Dopamine/administration & dosage , Drug Carriers/chemistry , Liposomes , Nanoparticles , Adhesiveness , Administration, Intranasal , Animals , Cells, Cultured , Chitosan/chemistry , Dopamine/pharmacokinetics , Dopamine/toxicity , Dopamine Agents/pharmacokinetics , Dopamine Agents/toxicity , Dose-Response Relationship, Drug , Lipids/chemistry , Mice , Olfactory Bulb/cytology , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Photoelectron Spectroscopy
16.
Science ; 372(6547): 1205-1209, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34112692

ABSTRACT

Quiescent neural stem cells (NSCs) in the adult mouse ventricular-subventricular zone (V-SVZ) undergo activation to generate neurons and some glia. Here we show that platelet-derived growth factor receptor beta (PDGFRß) is expressed by adult V-SVZ NSCs that generate olfactory bulb interneurons and glia. Selective deletion of PDGFRß in adult V-SVZ NSCs leads to their release from quiescence, uncovering gliogenic domains for different glial cell types. These domains are also recruited upon injury. We identify an intraventricular oligodendrocyte progenitor derived from NSCs inside the brain ventricles that contacts supraependymal axons. Together, our findings reveal that the adult V-SVZ contains spatial domains for gliogenesis, in addition to those for neurogenesis. These gliogenic NSC domains tend to be quiescent under homeostasis and may contribute to brain plasticity.


Subject(s)
Adult Stem Cells/physiology , Cerebral Ventricles/physiology , Lateral Ventricles/physiology , Neural Stem Cells/physiology , Neuroglia/physiology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Astrocytes/cytology , Astrocytes/physiology , Axons/physiology , Cell Differentiation , Cell Division , Cerebral Ventricles/cytology , Ependyma/cytology , Ependyma/physiology , Female , Gene Expression Profiling , Homeostasis , Lateral Ventricles/cytology , Male , Mice , Neurogenesis , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics
17.
Sci Immunol ; 6(60)2021 06 25.
Article in English | MEDLINE | ID: mdl-34172587

ABSTRACT

Viral encephalitis initiates a series of immunological events in the brain that can lead to brain damage and death. Astrocytes express IFN-ß in response to neurotropic infection, whereas activated microglia produce proinflammatory cytokines and accumulate at sites of infection. Here, we observed that neurotropic vesicular stomatitis virus (VSV) infection causes recruitment of leukocytes into the central nervous system (CNS), which requires MyD88, an adaptor of Toll-like receptor and interleukin-1 receptor signaling. Infiltrating leukocytes, and in particular CD8+ T cells, protected against lethal VSV infection of the CNS. Reconstitution of MyD88, specifically in neurons, restored chemokine production in the olfactory bulb as well as leukocyte recruitment into the infected CNS and enhanced survival. Comparative analysis of the translatome of neurons and astrocytes verified neurons as the critical source of chemokines, which regulated leukocyte infiltration of the infected brain and affected survival.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokines/metabolism , Encephalitis, Viral/immunology , Myeloid Differentiation Factor 88/metabolism , Rhabdoviridae Infections/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Disease Models, Animal , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Female , Humans , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Neurons/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/immunology , Olfactory Bulb/pathology , Olfactory Bulb/virology , Rhabdoviridae Infections/pathology , Rhabdoviridae Infections/virology , Signal Transduction/immunology , Vesiculovirus/immunology
18.
Cell Rep ; 35(12): 109276, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34161760

ABSTRACT

Developing neurons initially form excessive neurites and then remodel them based on molecular cues and neuronal activity. Developing mitral cells in the olfactory bulb initially extend multiple primary dendrites. They then stabilize single primary dendrites while eliminating others. However, the mechanisms underlying selective dendrite remodeling remain elusive. Using CRISPR-Cas9-based knockout screening combined with in utero electroporation, we identify BMPR-2 as a key regulator for selective dendrite stabilization. Bmpr2 knockout and its rescue experiments show that BMPR-2 inhibits LIMK without ligands and thereby permits dendrite destabilization. In contrast, the overexpression of antagonists and agonists indicates that ligand-bound BMPR-2 stabilizes dendrites, most likely by releasing LIMK. Using genetic and FRET imaging experiments, we demonstrate that free LIMK is activated by NMDARs via Rac1, facilitating dendrite stabilization through F-actin formation. Thus, the selective stabilization of primary dendrites is ensured by concomitant inputs of BMP ligands and neuronal activity.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Dendrites/metabolism , Olfactory Bulb/cytology , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type II/chemistry , Bone Morphogenetic Proteins/metabolism , Glutamic Acid/metabolism , Ligands , Lim Kinases/metabolism , Mice, Inbred C57BL , Protein Domains , Receptors, N-Methyl-D-Aspartate/metabolism , Structure-Activity Relationship , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
19.
J Neurosci ; 41(30): 6449-6467, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34099512

ABSTRACT

In sensory systems of the brain, mechanisms exist to extract distinct features from stimuli to generate a variety of behavioral repertoires. These often correspond to different cell types at various stages in sensory processing. In the mammalian olfactory system, complex information processing starts in the olfactory bulb, whose output is conveyed by mitral cells (MCs) and tufted cells (TCs). Despite many differences between them, and despite the crucial position they occupy in the information hierarchy, Cre-driver lines that distinguish them do not yet exist. Here, we sought to identify genes that are differentially expressed between MCs and TCs of the mouse, with an ultimate goal to generate a cell type-specific Cre-driver line, starting from a transcriptome analysis using a large and publicly available single-cell RNA-seq dataset (Zeisel et al., 2018). Many genes were differentially expressed, but only a few showed consistent expressions in MCs and at the specificity required. After further validating these putative markers using ISH, two genes (i.e., Pkib and Lbdh2) remained as promising candidates. Using CRISPR/Cas9-mediated gene editing, we generated Cre-driver lines and analyzed the resulting recombination patterns. This indicated that our new inducible Cre-driver line, Lbhd2-CreERT2, can be used to genetically label MCs in a tamoxifen dose-dependent manner, both in male and female mice, as assessed by soma locations, projection patterns, and sensory-evoked responses in vivo Hence, this is a promising tool for investigating cell type-specific contributions to olfactory processing and demonstrates the power of publicly accessible data in accelerating science.SIGNIFICANCE STATEMENT In the brain, distinct cell types play unique roles. It is therefore important to have tools for studying unique cell types specifically. For the sense of smell in mammals, information is processed first by circuits of the olfactory bulb, where two types of cells, mitral cells and tufted cells, output different information. We generated a transgenic mouse line that enables mitral cells to be specifically labeled or manipulated. This was achieved by looking for genes that are specific to mitral cells using a large and public gene expression dataset, and creating a transgenic mouse using the gene editing technique, CRISPR/Cas9. This will allow scientists to better investigate parallel information processing underlying the sense of smell.


Subject(s)
Cell Line , Neurons/cytology , Olfactory Bulb/cytology , Olfactory Perception/physiology , Animals , Female , Integrases , Male , Mice , Mice, Transgenic , Olfactory Pathways/cytology
20.
J Neurosci Res ; 99(7): 1835-1849, 2021 07.
Article in English | MEDLINE | ID: mdl-33960512

ABSTRACT

Spinal cord injury (SCI) is a debilitating condition, which leads to a permanent loss of functions below the injury site. The events which take place after SCI are characterized by cellular death, release of inhibitory factors, and inflammation. Many therapies have been studied to cure SCI, among them magnetic stimulation aims to reduce the secondary damages in particular by decreasing apoptosis, while, cellular transplantation promotes neuroregeneration by enhancing axonal regrowth. In the present study, we compared individually primary olfactory ensheathing cell (OEC) transplantation and repetitive trans-spinal magnetic stimulation (rTSMS) and then, we combined these two therapeutic approaches on tissue repair and functional recovery after SCI. To do so, SCIs were performed at Th10 level on female C57BL/6 mice, which were randomized into four groups: SCI, SCI + primary bOECs, SCI + STM, SCI + primary bulbar olfactory ensheathing cells (bOECs) + stimulation (STM). On these animals bioluminescence, immunohistological, and behavioral experiments were performed after SCI. Our results show that rTSMS has beneficial effect on the modulation of spinal scar by reducing fibrosis, demyelination, and microglial cell activation and by increasing the astroglial component of the scar, while, primary bOEC transplantation decreases microglial reactivity. At the opposite, locotronic experiments show that both treatments induce functional recovery. We did not observed any additional effect by combining the two therapeutic approaches. Taken together, the present study indicates that primary bOEC transplantation and rTSMS treatment act through different mechanisms after SCI to induce functional recovery. In our experimental paradigm, the combination of the two therapies does not induce any additional benefit.


Subject(s)
Cell Transplantation/methods , Neural Stem Cells/transplantation , Recovery of Function , Spinal Cord Injuries/pathology , Transcranial Magnetic Stimulation/methods , Animals , Female , Mice , Mice, Inbred C57BL , Nerve Regeneration , Olfactory Bulb/cytology , Random Allocation
SELECTION OF CITATIONS
SEARCH DETAIL
...