Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Nat Commun ; 15(1): 1877, 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38461182

ABSTRACT

Axonal growth cones mediate axonal guidance and growth regulation. We show that migrating neurons in mice possess a growth cone at the tip of their leading process, similar to that of axons, in terms of the cytoskeletal dynamics and functional responsivity through protein tyrosine phosphatase receptor type sigma (PTPσ). Migrating-neuron growth cones respond to chondroitin sulfate (CS) through PTPσ and collapse, which leads to inhibition of neuronal migration. In the presence of CS, the growth cones can revert to their extended morphology when their leading filopodia interact with heparan sulfate (HS), thus re-enabling neuronal migration. Implantation of an HS-containing biomaterial in the CS-rich injured cortex promotes the extension of the growth cone and improve the migration and regeneration of neurons, thereby enabling functional recovery. Thus, the growth cone of migrating neurons is responsive to extracellular environments and acts as a primary regulator of neuronal migration.


Subject(s)
Growth Cones , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Mice , Animals , Growth Cones/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Neurogenesis , Axons/metabolism , Chondroitin Sulfates/metabolism , Brain/metabolism , Cells, Cultured
2.
Biomaterials ; 294: 122003, 2023 03.
Article in English | MEDLINE | ID: mdl-36736095

ABSTRACT

The mammalian brain has very limited ability to regenerate lost neurons and recover function after injury. Promoting the migration of young neurons (neuroblasts) derived from endogenous neural stem cells using biomaterials is a new and promising approach to aid recovery of the brain after injury. However, the delivery of sufficient neuroblasts to distant injured sites is a major challenge because of the limited number of scaffold cells that are available to guide neuroblast migration. To address this issue, we have developed an amphiphilic peptide [(RADA)3-(RADG)] (mRADA)-tagged N-cadherin extracellular domain (Ncad-mRADA), which can remain in mRADA hydrogels and be injected into deep brain tissue to facilitate neuroblast migration. Migrating neuroblasts directly contacted the fiber-like Ncad-mRADA hydrogel and efficiently migrated toward an injured site in the striatum, a deep brain area. Furthermore, application of Ncad-mRADA to neonatal cortical brain injury efficiently promoted neuronal regeneration and functional recovery. These results demonstrate that self-assembling Ncad-mRADA peptides mimic both the function and structure of endogenous scaffold cells and provide a novel strategy for regenerative therapy.


Subject(s)
Cadherins , Neural Stem Cells , Animals , Brain , Neurons , Peptides , Mammals
3.
Stem Cell Reports ; 17(12): 2704-2717, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36368330

ABSTRACT

The concept of a perivascular niche has been proposed for neural stem cells (NSCs). This study examined endothelial colony-forming cell (ECFC)-secreted proteins as potential niche factors for NSCs. Intraventricle infusion with ECFC-secreted proteins increased the number of NSCs. ECFC-secreted proteins were more effective in promoting NSC self-renewal than marrow stromal cell (MSC)-secreted proteins. Differential proteomics analysis of MSC-secreted and ECFC-secreted proteins was performed, which revealed chitinase-like protein 3 (CHIL3; also called ECF-L or Ym1) as a candidate niche factor for NSCs. Experiments with recombinant CHIL3, small interfering RNA, and neutralizing antibodies demonstrated that CHIL3 stimulated NSC self-renewal with neurogenic propensity. CHIL3 was endogenously expressed in the neurogenic niche of the brain and retina as well as in the injured brain and retina. Transcriptome and phosphoproteome analyses revealed that CHIL3 activated various genes and proteins associated with NSC maintenance or neurogenesis. Thus, CHIL3 is a novel niche factor for NSCs.


Subject(s)
Chitinases , Neural Stem Cells , Animals , Mice , Stem Cell Niche , Chitinases/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Brain/metabolism
5.
Nat Commun ; 12(1): 6623, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799548

ABSTRACT

During injured tissue regeneration, the extracellular matrix plays a key role in controlling and coordinating various cellular events by binding and releasing secreted proteins in addition to promoting cell adhesion. Herein, we develop a cell-adhesive fiber-forming peptide that mimics the jigsaw-shaped hydrophobic surface in the dovetail-packing motif of glycophorin A as an artificial extracellular matrix for regenerative therapy. We show that the jigsaw-shaped self-assembling peptide forms several-micrometer-long supramolecular nanofibers through a helix-to-strand transition to afford a hydrogel under physiological conditions and disperses homogeneously in the hydrogel. The molecular- and macro-scale supramolecular properties of the jigsaw-shaped self-assembling peptide hydrogel allow efficient incorporation and sustained release of vascular endothelial growth factor, and demonstrate cell transplantation-free regenerative therapeutic effects in a subacute-chronic phase mouse stroke model. This research highlights a therapeutic strategy for injured tissue regeneration using the jigsaw-shaped self-assembling peptide supramolecular hydrogel.


Subject(s)
Brain Regeneration/physiology , Hydrogels/chemistry , Peptides/chemistry , Proteins/chemistry , Adhesives , Animals , Biomedical Engineering , Brain Injuries/diagnostic imaging , Cell Adhesion , Disease Models, Animal , Female , Green Fluorescent Proteins/chemistry , Hydrogels/therapeutic use , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Inbred C57BL , Nanofibers , Nervous System , Peptides/therapeutic use , Vascular Endothelial Growth Factor A
6.
Sci Transl Med ; 13(587)2021 03 31.
Article in English | MEDLINE | ID: mdl-33790026

ABSTRACT

The lateral ventricle (LV) is flanked by the subventricular zone (SVZ), a neural stem cell (NSC) niche rich in extrinsic growth factors regulating NSC maintenance, proliferation, and neuronal differentiation. Dysregulation of the SVZ niche causes LV expansion, a condition known as hydrocephalus; however, the underlying pathological mechanisms are unclear. We show that deficiency of the proteoglycan Tsukushi (TSK) in ependymal cells at the LV surface and in the cerebrospinal fluid results in hydrocephalus with neurodevelopmental disorder-like symptoms in mice. These symptoms are accompanied by altered differentiation and survival of the NSC lineage, disrupted ependymal structure, and dysregulated Wnt signaling. Multiple TSK variants found in patients with hydrocephalus exhibit reduced physiological activity in mice in vivo and in vitro. Administration of wild-type TSK protein or Wnt antagonists, but not of hydrocephalus-related TSK variants, in the LV of TSK knockout mice prevented hydrocephalus and preserved SVZ neurogenesis. These observations suggest that TSK plays a crucial role as a niche molecule modulating the fate of SVZ NSCs and point to TSK as a candidate for the diagnosis and therapy of hydrocephalus.


Subject(s)
Hydrocephalus , Neural Stem Cells , Neurogenesis , Proteoglycans , Animals , Cell Proliferation , Humans , Mice , Mice, Knockout , Stem Cell Niche
7.
Mol Brain ; 14(1): 66, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33832520

ABSTRACT

GAP-43 is a vertebrate neuron-specific protein and that is strongly related to axon growth and regeneration; thus, this protein has been utilized as a classical molecular marker of these events and growth cones. Although GAP-43 was biochemically characterized more than a quarter century ago, how this protein is related to these events is still not clear. Recently, we identified many phosphorylation sites in the growth cone membrane proteins of rodent brains. Two phosphorylation sites of GAP-43, S96 and T172, were found within the top 10 hit sites among all proteins. S96 has already been characterized (Kawasaki et al., 2018), and here, phosphorylation of T172 was characterized. In vitro (cultured neurons) and in vivo, an antibody specific to phosphorylated T172 (pT172 antibody) specifically recognized cultured growth cones and growing axons in developing mouse neurons, respectively. Immunoblotting showed that pT172 antigens were more rapidly downregulated throughout development than those of pS96 antibody. From the primary structure, this phosphorylation site was predicted to be conserved in a wide range of animals including primates. In the developing marmoset brainstem and in differentiated neurons derived from human induced pluripotent stem cells, immunoreactivity with pT172 antibody revealed patterns similar to those in mice. pT172 antibody also labeled regenerating axons following sciatic nerve injury. Taken together, the T172 residue is widely conserved in a wide range of mammals including primates, and pT172 is a new candidate molecular marker for growing axons.


Subject(s)
Axons/metabolism , Biomarkers/metabolism , GAP-43 Protein/metabolism , Mammals/metabolism , Phosphothreonine/metabolism , Amino Acid Sequence , Animals , Antibodies/metabolism , Brain/embryology , Callithrix , Cells, Cultured , Ferrets , GAP-43 Protein/chemistry , Growth Cones/metabolism , HEK293 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Nerve Regeneration , Phosphorylation , Primates , Sciatic Nerve/injuries
8.
Mol Brain ; 14(1): 30, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33568175

ABSTRACT

Metabolites underlying brain function and pathology are not as well understood as genes. Here, we applied a novel metabolomics approach to further understand the mechanisms of memory processing in sleep. As hippocampal dentate gyrus neurons are known to consolidate contextual fear memory, we analyzed real-time changes in metabolites in the dentate gyrus in different sleep-wake states in mice. Throughout the study, we consistently detected more than > 200 metabolites. Metabolite profiles changed dramactically upon sleep-wake state transitions, leading to a clear separation of phenotypes between wakefulness and sleep. By contrast, contextual fear memory consolidation induced less obvious metabolite phenotypes. However, changes in purine metabolites were observed upon both sleep-wake state transitions and contextual fear memory consolidation. Dietary supplementation of certain purine metabolites impaired correlations between conditioned fear responses before and after memory consolidation. These results point toward the importance of purine metabolism in fear memory processing during sleep.


Subject(s)
Fear/physiology , Memory Consolidation/physiology , Metabolomics , Sleep/physiology , Administration, Oral , Animals , Mice, Inbred C57BL , Purines/administration & dosage , Purines/metabolism , Wakefulness/physiology
9.
Neurosci Res ; 167: 64-69, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32553727

ABSTRACT

The ventricular-subventricular zone (V-SVZ) is located in the walls of the lateral ventricles and produces new neurons in the postnatal brain of mammals, including humans. Immature new neurons called "neuroblasts" generated by neural stem cells in the V-SVZ migrate toward their final destinations and contribute to brain development and plasticity. In this review, we describe recent progress in understanding the similarities and dissimilarities in postnatal neurogenesis and neuronal migration between rodents and primates. In rodents, most new V-SVZ-derived neurons migrate along the rostral migratory stream towards the olfactory bulb, where they differentiate into interneurons. In contrast, in humans, the extensive migration of new neurons towards the neocortex continues for several months after birth and might be involved in the development of the expanded neocortex. The mode of migration and the fate of neuroblasts seem to change depending on their environment, destination, and roles in the brain. A better understanding of these similarities and differences between rodents and primates will help translate important findings from animal models and may contribute to the development of clinical strategies for brain repair.


Subject(s)
Lateral Ventricles , Rodentia , Animals , Cell Movement , Neurogenesis , Olfactory Bulb , Primates
10.
Mol Brain ; 13(1): 98, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32590991

ABSTRACT

In many mammalian species, the production of new neurons in the hippocampal dentate gyrus continues throughout life. Previous studies using rodents suggest that adult-born neurons are involved in memory and cognition tasks and mood regulation. Interferon-alpha (IFNα), a proinflammatory cytokine used for the treatment of chronic viral hepatitis and malignancies, frequently causes depressive symptoms in patients and animals, including non-human primates. We have previously demonstrated that chronic IFNα treatment decreases hippocampal neurogenesis in mice. Here, we investigated the effects of four-week human pegylated IFNα treatment on hippocampal neurogenesis and behavior in common marmosets. Continuous monitoring of voluntary activity levels using an actigraphy device suggested that adaptive ability is impaired in IFNα-treated animals. Analyses of BrdU-labeled cells expressing a marker for immature or mature neurons revealed a significant reduction in the number of new neurons in the hippocampus of IFNα-treated animals. These data indicate that chronic human IFNα treatment causes behavioral changes and a decrease in hippocampal neurogenesis in common marmosets.


Subject(s)
Behavior, Animal/physiology , Hippocampus/physiology , Interferon-alpha/pharmacology , Neurogenesis/drug effects , Animals , Behavior, Animal/drug effects , Callithrix , Female , Hippocampus/drug effects , Humans , Male
11.
Neuron ; 107(3): 552-565.e10, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32502462

ABSTRACT

The occurrence of dreaming during rapid eye movement (REM) sleep prompts interest in the role of REM sleep in hippocampal-dependent episodic memory. Within the mammalian hippocampus, the dentate gyrus (DG) has the unique characteristic of exhibiting neurogenesis persisting into adulthood. Despite their small numbers and sparse activity, adult-born neurons (ABNs) in the DG play critical roles in memory; however, their memory function during sleep is unknown. Here, we investigate whether young ABN activity contributes to memory consolidation during sleep using Ca2+ imaging in freely moving mice. We found that contextual fear learning recruits a population of young ABNs that are reactivated during subsequent REM sleep against a backdrop of overall reduced ABN activity. Optogenetic silencing of this sparse ABN activity during REM sleep alters the structural remodeling of spines on ABN dendrites and impairs memory consolidation. These findings provide a causal link between ABN activity during REM sleep and memory consolidation.


Subject(s)
Conditioning, Psychological , Dentate Gyrus/physiology , Memory Consolidation/physiology , Neurons/physiology , Sleep, REM/physiology , Animals , Calcium/metabolism , Dentate Gyrus/cytology , Electroencephalography , Electromyography , Fear , Hippocampus , Learning , Mice , Neurogenesis , Optogenetics , Theta Rhythm
12.
Cereb Cortex ; 30(7): 4092-4109, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32108222

ABSTRACT

Even after birth, neuronal production continues in the ventricular-subventricular zone (V-SVZ) and hippocampus in many mammals. The immature new neurons ("neuroblasts") migrate and then mature at their final destination. In humans, neuroblast production and migration toward the neocortex and the olfactory bulb (OB) occur actively only for a few months after birth and then sharply decline with age. However, the precise spatiotemporal profiles and fates of postnatally born neurons remain unclear due to methodological limitations. We previously found that common marmosets, small nonhuman primates, share many features of V-SVZ organization with humans. Here, using marmosets injected with thymidine analogue(s) during various postnatal periods, we demonstrated spatiotemporal changes in neurogenesis during development. V-SVZ progenitor proliferation and neuroblast migration toward the OB and neocortex sharply decreased by 4 months, most strikingly in a V-SVZ subregion from which neuroblasts migrated toward the neocortex. Postnatally born neurons matured within a few months in the OB and hippocampus but remained immature until 6 months in the neocortex. While neurogenic activity was sustained for a month after birth, the distribution and/or differentiation diversity was more restricted in 1-month-born cells than in the neonatal-born population. These findings shed light on distinctive features of postnatal neurogenesis in primates.


Subject(s)
Cell Proliferation , Hippocampus/growth & development , Lateral Ventricles/growth & development , Neocortex/growth & development , Neural Stem Cells/cytology , Neurogenesis , Olfactory Bulb/growth & development , Animals , Brain/cytology , Brain/growth & development , Callithrix , Cell Movement , Cerebral Ventricles/cytology , Cerebral Ventricles/growth & development , Hippocampus/cytology , Lateral Ventricles/cytology , Neocortex/cytology , Olfactory Bulb/cytology , Spatio-Temporal Analysis
13.
J Neurosci ; 39(50): 9967-9988, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31685650

ABSTRACT

New neurons, referred to as neuroblasts, are continuously generated in the ventricular-subventricular zone of the brain throughout an animal's life. These neuroblasts are characterized by their unique potential for proliferation, formation of chain-like cell aggregates, and long-distance and high-speed migration through the rostral migratory stream (RMS) toward the olfactory bulb (OB), where they decelerate and differentiate into mature interneurons. The dynamic changes of ultrastructural features in postnatal-born neuroblasts during migration are not yet fully understood. Here we report the presence of a primary cilium, and its ultrastructural morphology and spatiotemporal dynamics, in migrating neuroblasts in the postnatal RMS and OB. The primary cilium was observed in migrating neuroblasts in the postnatal RMS and OB in male and female mice and zebrafish, and a male rhesus monkey. Inhibition of intraflagellar transport molecules in migrating neuroblasts impaired their ciliogenesis and rostral migration toward the OB. Serial section transmission electron microscopy revealed that each migrating neuroblast possesses either a pair of centrioles or a basal body with an immature or mature primary cilium. Using immunohistochemistry, live imaging, and serial block-face scanning electron microscopy, we demonstrate that the localization and orientation of the primary cilium are altered depending on the mitotic state, saltatory migration, and deceleration of neuroblasts. Together, our results highlight a close mutual relationship between spatiotemporal regulation of the primary cilium and efficient chain migration of neuroblasts in the postnatal brain.SIGNIFICANCE STATEMENT Immature neurons (neuroblasts) generated in the postnatal brain have a mitotic potential and migrate in chain-like cell aggregates toward the olfactory bulb. Here we report that migrating neuroblasts possess a tiny cellular protrusion called a primary cilium. Immunohistochemical studies with zebrafish, mouse, and monkey brains suggest that the presence of the primary cilium in migrating neuroblasts is evolutionarily conserved. Ciliogenesis in migrating neuroblasts in the rostral migratory stream is suppressed during mitosis and promoted after cell cycle exit. Moreover, live imaging and 3D electron microscopy revealed that ciliary localization and orientation change during saltatory movement of neuroblasts. Our results reveal highly organized dynamics in maturation and positioning of the primary cilium during neuroblast migration that underlie saltatory movement of postnatal-born neuroblasts.


Subject(s)
Cell Movement/physiology , Cilia/ultrastructure , Lateral Ventricles/ultrastructure , Neural Stem Cells/ultrastructure , Neurons/ultrastructure , Olfactory Bulb/ultrastructure , Animals , Female , Macaca mulatta , Male , Mice , Zebrafish
14.
Neurochem Int ; 126: 69-73, 2019 06.
Article in English | MEDLINE | ID: mdl-30851365

ABSTRACT

Neurogenesis and angiogenesis share regulatory factors that contribute to the formation of vascular networks and neuronal circuits in the brain. While crosstalk mechanisms between neural stem cells (NSCs) and the vasculature have been extensively investigated, recent studies have provided evidence that blood vessels also play an essential role in neuronal migration in the brain during development and regeneration. The mechanisms of the neuronal migration along blood vessels, referred to as "vascular-guided migration," are now being elucidated. The vascular endothelial cells secrete soluble factors that attract and promote neuronal migration in collaboration with astrocytes that enwrap the blood vessels. In addition, especially in the adult brain, the blood vessels serve as a migration scaffold for adult-born immature neurons generated in the ventricular-subventricular zone (V-SVZ), a germinal zone surrounding the lateral ventricles. The V-SVZ-derived immature neurons use the vascular scaffold to assist their migration toward an injured area after ischemic stroke, and contribute to neuronal regeneration. Here we review the current knowledge about the role of vasculature in neuronal migration and the molecular mechanisms controlling this process. While most of this research has been done in rodents, a comprehensive understanding of vasculature-guided neuronal migration could contribute to new therapeutic approaches for increasing new neurons in the brain after injury.


Subject(s)
Blood Vessels/physiology , Brain/physiology , Cell Movement/physiology , Neural Stem Cells/physiology , Neurons/physiology , Tissue Scaffolds/trends , Animals , Astrocytes/physiology , Blood Vessels/cytology , Blood-Brain Barrier/cytology , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/cytology , Humans , Neurogenesis/physiology , Stroke/pathology , Stroke/therapy
15.
Mol Biol Cell ; 30(1): 56-68, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30379609

ABSTRACT

Neural stem cells (NSCs) are retained in the adult ventricular-subventricular zone (V-SVZ), a specialized neurogenic niche with a unique cellular architecture. It currently remains unclear whether or how NSCs utilize basement membranes (BMs) in this niche. Here, we examine the molecular compositions and functions of BMs in the adult mouse V-SVZ. Whole-mount V-SVZ immunostaining revealed that fractones, which are fingerlike processes of extravascular BMs, are speckled BMs unconnected to the vasculature, and differ in their molecular composition from vascular BMs. Glial fibrillary acidic protein (GFAP)-positive astrocytes and NSCs produce and adhere to speckled BMs. Furthermore, Gfap-Cre-mediated Lamc1flox(E1605Q) knockin mice, in which integrin-binding activities of laminins are specifically nullified in GFAP-positive cells, exhibit a decreased number and size of speckled BMs and reduced in vitro neurosphere-forming activity. Our results reveal niche activities of fractones/speckled BMs for NSCs and provide molecular insights into how laminin-integrin interactions regulate NSCs in vivo.


Subject(s)
Basement Membrane/metabolism , Lateral Ventricles/metabolism , Neural Stem Cells/metabolism , Stem Cell Niche , Animals , Animals, Newborn , Basement Membrane/blood supply , Basement Membrane/cytology , Cell Proliferation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ependyma/cytology , Ependyma/metabolism , Glial Fibrillary Acidic Protein/metabolism , Integrins/metabolism , Laminin/metabolism , Lateral Ventricles/cytology , Mice, Inbred C57BL , Mutation/genetics , Neural Stem Cells/cytology
16.
Cell Rep ; 25(3): 624-639.e6, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332643

ABSTRACT

As an essential step for brain morphogenesis, neurons migrate via mechanical interactions with components of their environment such as neighboring cells and the extracellular matrix. However, the molecular mechanism by which neurons exert forces on their environment during migration remains poorly understood. Here, we show that shootin1b is expressed in migrating mouse olfactory interneurons and accumulates at their leading process growth cone. We demonstrate that shootin1b, by binding to cortactin and L1-CAM, couples F-actin retrograde flow and the adhesive substrate as a clutch molecule. Shootin1b-mediated clutch coupling at the growth cone generates traction force on the substrate, thereby promoting leading process extension and subsequent somal translocation of olfactory interneurons. Furthermore, loss of shootin1 causes abnormal positioning of the interneurons and dysgenesis of the olfactory bulb. Our findings indicate that shootin1b plays a key role in force-driven leading process extension, which propels the migration of olfactory interneurons during olfactory bulb formation.


Subject(s)
Brain/physiology , Cell Movement , Growth Cones/physiology , Interneurons/physiology , Nerve Tissue Proteins/physiology , Olfactory Bulb/physiology , Actins/metabolism , Animals , Brain/cytology , Cell Adhesion , Cells, Cultured , Female , Interneurons/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Olfactory Bulb/cytology , Rats, Wistar
17.
Cell Stem Cell ; 22(6): 783-784, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29859165

ABSTRACT

Adult neural stem cells in the wall of brain ventricles make direct contact with cerebrospinal fluid. In this issue of Cell Stem Cell, Petrik et al. (2018) demonstrate that these neural stem cells sense the flow of cerebrospinal fluid through a transmembrane sodium channel, ENaC, which regulates their proliferation.


Subject(s)
Adult Stem Cells , Neural Stem Cells , Adult , Cell Proliferation , Cerebral Ventricles , Epithelial Sodium Channels , Humans
18.
J Dermatol ; 45(8): 963-970, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29897137

ABSTRACT

Little attention has been given to the burden of chronic urticaria (CU) in Japan compared with other skin diseases, such as atopic dermatitis (AD) and psoriasis. The primary objective of the RELEASE study was to evaluate the real-life quality-of-life impairment in CU patients in Japan. Data were collected from 1443 urticaria, 1668 AD and 435 psoriatic patients; 552 urticaria patients who presented urticaria symptoms for over 6 weeks were defined as CU. The mean Dermatology Life Quality Index (DLQI) total score was 4.8, 6.1 and 4.8 in CU, AD and psoriatic patients, respectively. Disease control of urticaria evaluated by the Urticaria Control Test (UCT) and DLQI exhibited a strong correlation with a Spearman's rank correlation coefficient of -0.7158. CU and AD patients had relatively higher scores in all Work Productivity and Activity Impairment - General Health subscales except for absenteeism. At the time of the survey, approximately 64% of CU patients reported UCT scores of <12 and demonstrated higher work productivity loss and activity impairment versus patients with UCT scores of ≥12. Patients with lower UCT scores also displayed a higher percentage of dissatisfaction with their health state and the treatment they received. Approximately 85% of patients with CU had visited dermatology clinics, and less than 20% had visited hospital, indicating existence of a highly burdened population outside specialized centers. These results highlight the unmet medical needs of CU patients, suggesting the need to increase awareness of CU burden among both physicians and patients and to pursue improved real-life patient care.


Subject(s)
Cost of Illness , Health Status , Patient Satisfaction/statistics & numerical data , Quality of Life , Urticaria/complications , Absenteeism , Adult , Chronic Disease , Dermatitis, Atopic/complications , Dermatitis, Atopic/epidemiology , Dermatitis, Atopic/physiopathology , Dermatitis, Atopic/therapy , Efficiency , Female , Health Services Needs and Demand/statistics & numerical data , Humans , Japan/epidemiology , Male , Middle Aged , Patient Reported Outcome Measures , Prevalence , Psoriasis/complications , Psoriasis/epidemiology , Psoriasis/physiopathology , Psoriasis/therapy , Urticaria/epidemiology , Urticaria/physiopathology , Urticaria/therapy
19.
J Neurosci ; 38(19): 4598-4609, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29661967

ABSTRACT

In the rodent olfactory system, neuroblasts produced in the ventricular-subventricular zone of the postnatal brain migrate tangentially in chain-like cell aggregates toward the olfactory bulb (OB) through the rostral migratory stream (RMS). After reaching the OB, the chains are dissociated and the neuroblasts migrate individually and radially toward their final destination. The cellular and molecular mechanisms controlling cell-cell adhesion during this detachment remain unclear. Here we report that Fyn, a nonreceptor tyrosine kinase, regulates the detachment of neuroblasts from chains in the male and female mouse OB. By performing chemical screening and in vivo loss-of-function and gain-of-function experiments, we found that Fyn promotes somal disengagement from the chains and is involved in neuronal migration from the RMS into the granule cell layer of the OB. Fyn knockdown or Dab1 (disabled-1) deficiency caused p120-catenin to accumulate and adherens junction-like structures to be sustained at the contact sites between neuroblasts. Moreover, a Fyn and N-cadherin double-knockdown experiment indicated that Fyn regulates the N-cadherin-mediated cell adhesion between neuroblasts. These results suggest that the Fyn-mediated control of cell-cell adhesion is critical for the detachment of chain-forming neuroblasts in the postnatal OB.SIGNIFICANCE STATEMENT In the postnatal brain, newly born neurons (neuroblasts) migrate in chain-like cell aggregates toward their destination, where they are dissociated into individual cells and mature. The cellular and molecular mechanisms controlling the detachment of neuroblasts from chains are not understood. Here we show that Fyn, a nonreceptor tyrosine kinase, promotes the somal detachment of neuroblasts from chains, and that this regulation is critical for the efficient migration of neuroblasts to their destination. We further show that Fyn and Dab1 (disabled-1) decrease the cell-cell adhesion between chain-forming neuroblasts, which involves adherens junction-like structures. Our results suggest that Fyn-mediated regulation of the cell-cell adhesion of neuroblasts is critical for their detachment from chains in the postnatal brain.


Subject(s)
Brain/physiology , Neural Stem Cells/physiology , Proto-Oncogene Proteins c-fyn/physiology , Animals , Brain/cytology , Brain/growth & development , Cadherins/genetics , Catenins/metabolism , Cell Adhesion/physiology , Cell Movement/genetics , Female , Gene Knockdown Techniques , Male , Mice , Nerve Tissue Proteins/genetics , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , Olfactory Bulb/physiology
20.
Cell Stem Cell ; 22(1): 128-137.e9, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29276142

ABSTRACT

Radial glia (RG) are embryonic neural stem cells (NSCs) that produce neuroblasts and provide fibers that act as a scaffold for neuroblast migration during embryonic development. Although they normally disappear soon after birth, here we found that RG fibers can persist in injured neonatal mouse brains and act as a scaffold for postnatal ventricular-subventricular zone (V-SVZ)-derived neuroblasts that migrate to the lesion site. This injury-induced maintenance of RG fibers has a limited time window during post-natal development and promotes directional saltatory movement of neuroblasts via N-cadherin-mediated cell-cell contacts that promote RhoA activation. Transplanting an N-cadherin-containing scaffold into injured neonatal brains likewise promotes migration and maturation of V-SVZ-derived neuroblasts, leading to functional improvements in impaired gait behaviors. Together these results suggest that RG fibers enable postnatal V-SVZ-derived neuroblasts to migrate toward sites of injury, thereby enhancing neuronal regeneration and functional recovery from neonatal brain injuries.


Subject(s)
Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Movement , Neuroglia/pathology , Neurons/pathology , Recovery of Function , Animals , Animals, Newborn , Cadherins/metabolism , Lateral Ventricles/pathology , Neuroglia/metabolism , Neuroglia/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...