Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters











Publication year range
1.
Front Neurosci ; 18: 1418058, 2024.
Article in English | MEDLINE | ID: mdl-39176381

ABSTRACT

Desmoplakin (Dsp) is a component of desmosomal cell-cell junctions that interacts with the cadherin complex and cytoskeletal intermediate filaments. In addition to its function as an adhesion component, Dsp is involved in various biological processes, such as gene expression, differentiation, and migration. Dsp is specifically expressed in the hippocampal dentate gyrus (DG) in the central nervous system. However, it is unclear how Dsp impacts hippocampal function and its related behaviors. Using an adeno-associated virus knockdown system in mice, we provide evidence that Dsp in the DG maintains hippocampal functions, including neuronal activity and adult neurogenesis, and contributes to anxiolytic-like effects. Dsp protein is mostly localized in mature granule cells in the adult DG. Dsp knockdown in the DG resulted in a lowered expression of an activity-dependent transcription factor FosB, and an increased expression of mature neuronal markers, such as calbindin. In addition, the suppression of Dsp decreases serotonin responsiveness at the DG output mossy fiber synapses and alters adult neurogenic processes in the subgranular zone of the DG. Moreover, DG-specific Dsp knockdown mice showed an increase in anxiety-like behaviors. Taken together, this research uncovers an unexplored function for Dsp in the central nervous system and suggests that Dsp in the DG may function as a regulator to maintain proper neuronal activation and adult neurogenesis, and contribute to the adaptation of emotion-related behavior.

2.
Int J Surg Case Rep ; 119: 109737, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38714068

ABSTRACT

INTRODUCTION AND IMPORTANCE: Although bilateral congenital choanal atresia (CCA) requires early intervention to open closure walls for safe breathing, it is desirable to be withheld until an infant acquires surgical and anesthetic tolerance. Here we introduce an infant of CCA whose closure wall had thickened during a waiting period for an elective surgery. CASE PRESENTATION: The choana of the patient could not be identified by intranasal fiberscopy and the bilateral CCA was found by CT scan on day 17 after birth. Since he could breathe orally without distress, surgery was withheld until he acquires the tolerance. At nine weeks old, however, CT image detected thickening of the closure wall. At 10 weeks old, he underwent scheduled surgery in which the bilateral closure walls were removed together with attached posterior part of the nasal septum under endoscopic endonasal approach. The patient became able to breath nasally and the choana remained open without restenosis at 3 years after surgery. CLINICAL DISCUSSION: This is the first CCA case reporting closure walls thickened during a waiting period for an elective surgery. Although waiting for surgery was systemically safer by growth, the surgery became more invasive to prevention from restenosis. CONCLUSIONS: This case suggests that we must decide appropriate timing of surgery in an infant, considering dilemma between systemic safety ensuring and lesion aggravation by waiting for surgery.

3.
Otol Neurotol ; 45(2): e102-e106, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38013495

ABSTRACT

OBJECTIVE: To evaluate pneumatization and opacification of the temporal bone on computed tomography (CT) images in patients with primary ciliary dyskinesia (PCD). STUDY DESIGN: Retrospective case-control study. SETTING: Tertiary referral center. PATIENTS: Fifteen patients with PCD (30 ears) and 45 age-matched individuals without PCD (90 ears) as controls. INTERVENTION: Diagnostic only. MAIN OUTCOME MEASURES: Quantification of mastoid air cells in the PCD and control groups and comparison between them. Degree of middle ear opacification on CT images of the temporal bone in the PCD group. RESULTS: The volume of the mastoid air cells was 30% smaller in the PCD group than in the control group ( p < 0.05). The suppression ratio, which is defined to indicate how much the average volume of mastoid air cells in the PCD group is suppressed relative to the control group, was 64% lower in the PCD group ( p < 0.05). Opacification was noted in 47% of the mastoid air cells and 63% of the tympanic cavity on CT images of the temporal bone in the PCD group, which were significantly higher frequencies than in the control group (1.1% and 1.1%, respectively). CONCLUSIONS: Compared with individuals without PCD, those with PCD showed a significantly smaller volume of mastoid air cells and a significantly higher frequency of opacification of mastoid air cells and tympanic cavity on temporal bone CT. Otitis media raises suspicion for PCD, and the otological manifestations of PCD reported here could help to narrow the differential diagnosis and facilitate early treatment.


Subject(s)
Ciliary Motility Disorders , Mastoid , Humans , Mastoid/diagnostic imaging , Retrospective Studies , Case-Control Studies , Ear, Middle/diagnostic imaging
4.
Biol Pharm Bull ; 46(9): 1176-1183, 2023.
Article in English | MEDLINE | ID: mdl-37661396

ABSTRACT

Neuroinflammation is often associated with the development of depressive and anxiety disorders. The hippocampus is one of the brain regions affected by inflammation that is associated with these symptoms. However, the mechanism of hippocampal inflammation-induced emotional behavior remains unknown. The aim of this study was to clarify temporal changes in the neuroinflammatory responses in the hippocampus and the response of dentate gyrus (DG) neurons using peripheral lipopolysaccharide (LPS)-challenged mice. LPS administration induced anxiety-like activity in the elevated plus maze test and social interaction test after 24 h, at which time the mice had recovered from sickness behavior. We examined the hippocampal inflammation-related gene expression changes over time. The expression of interleukin-1ß (Il1b) and tumor necrosis factor α (Tnfa) was rapidly enhanced and sustained until 24 h after LPS administration, whereas the expression of Il6 was transiently induced at approx. 6 h. IL-6-dependent downstream signaling of transducer and activator of transcription 3 (STAT3) was also activated approx. 3-6 h after LPS treatment. The expression of innate immune genes including interferon-induced transmembrane proteins such as interferon-induced transmembrane protein 1 (Ifitm1) and Ifitm3 and complement factors such as C1qa and C1qb started to increase approx. 6 h and showed sustained or further increase at 24 h. We also examined changes in the expression of several maturation markers in the DG and found that LPS enhanced the expression of calbindin 1 (Calb1), tryptophan-2,3-dioxigenase 2 (Tdo2), Il1rl, and neurotrophin-3 (Ntf3) at 24 h after LPS treatment. Collectively, these results demonstrate temporal changes of inflammation and gene expression in the hippocampus in LPS-induced sickness and anxiety-like behaviors.


Subject(s)
Anxiety , Lipopolysaccharides , Animals , Mice , Lipopolysaccharides/toxicity , Anxiety/chemically induced , Anxiety/genetics , Inflammation/chemically induced , Inflammation/genetics , Hippocampus , Interferons , Gene Expression
5.
Front Neurosci ; 17: 1178555, 2023.
Article in English | MEDLINE | ID: mdl-37575306

ABSTRACT

The dentate gyrus (DG) of the hippocampus regulates stress-related emotional behaviors and ensures neurogenesis throughout life. Neurotrophin-3 (NT-3) is a neurotrophic factor that regulates neuronal differentiation, survival, and synaptic formation in both the peripheral and central nervous systems. NT-3 is expressed in the adult DG of the hippocampus; several chronic stress conditions enhance NT-3 expression in rodents. However, functional modulation of the adult DG by NT-3 signaling remains unclear. To directly investigate the impact of NT-3 on DG function, NT-3 was overexpressed in the hippocampal ventral DG by an adeno-associated virus carrying NT-3 (AAV-NT-3). Four weeks following the AAV-NT-3 injection, high NT-3 expression was observed in the ventral DG. We examined the influence of NT-3 overexpression on the neuronal responses and neurogenic processes in the ventral DG. NT-3 overexpression significantly increased the expression of the mature DG neuronal marker calbindin and immediate early genes, such as Fos and Fosb, thereby suggesting DG neuronal activation. During neurogenesis, the number of proliferating cells and immature neurons in the subgranular zone of the DG significantly decreased in the AAV-NT-3 group. Among the neurogenesis-related factors, Vegfd, Lgr6, Bmp7, and Drd1 expression significantly decreased. These results demonstrated that high NT-3 levels in the hippocampus regulate the activation of mature DG neurons and suppress the early phase of neurogenic processes, suggesting a possible role of NT-3 in the regulation of adult hippocampal function under stress conditions.

6.
Neuropharmacology ; 232: 109511, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37001727

ABSTRACT

Delta opioid receptors (DOPs) play an important role in depression and other mood disorders. However, little is known about the underlying physiological mechanisms. The hypothalamic-pituitary-adrenal axis, adult hippocampal neurogenesis, and neuroinflammation are regarded as key pathophysiological factors in depression. In this study, we investigated the influence of DOP activation on those factors in a valid animal model of depression, chronic vicarious social defeat stress (cVSDS) mice. cVSDS mice (male C57BL/6J mice) were produced following a 10-day exposure to witness of social defeat stress, and each evaluation was performed more than 28 days after the stress period. Repeated administrations to cVSDS mice with a selective DOP agonist, KNT-127, both during (10 days) and after (28 days) the stress period respectively improved their decreased social interaction behaviors and increased serum corticosterone levels. When administered during the stress period, KNT-127 suppressed decreases in the hippocampal newborn neuron survival rate in cVSDS mice. Moreover, in both administration paradigms, KNT-127 reduced the number of Iba-1- and CD11b-positive cells in the subgranular zone and the granule cell layer of the hippocampal dentate gyrus, indicating a suppression of cVSDS-induced microglial overactivation. These results suggest that KNT-127 acts over the hypothalamic-pituitary-adrenal axis and regulates neurogenesis and neuroinflammation resulting in anti-stress effects, and the antidepressant-like effects of the DOP agonist are implicated in the suppression of the neuroinflammation. This study presents a new finding on the effects of repeated DOP activations on the pathophysiological states of depression.


Subject(s)
Receptors, Opioid, delta , Social Defeat , Male , Mice , Animals , Receptors, Opioid, delta/agonists , Hypothalamo-Hypophyseal System/metabolism , Neuroinflammatory Diseases , Mice, Inbred C57BL , Pituitary-Adrenal System/metabolism , Analgesics, Opioid/pharmacology , Hippocampus , Dentate Gyrus/metabolism , Stress, Psychological/drug therapy , Neurogenesis , Depression/drug therapy
7.
Proc Natl Acad Sci U S A ; 119(33): e2117903119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35939697

ABSTRACT

Dopamine D1 receptors (D1Rs) in the hippocampal dentate gyrus (DG) are essential for antidepressant effects. However, the midbrain dopaminergic neurons, the major source of dopamine in the brain, only sparsely project to DG, suggesting possible activation of DG D1Rs by endogenous substances other than dopamine. We have examined this possibility using electrophysiological and biochemical techniques and found robust activation of D1Rs in mouse DG neurons by noradrenaline. Noradrenaline at the micromolar range potentiated synaptic transmission at the DG output and increased the phosphorylation of protein kinase A substrates in DG via activation of D1Rs and ß adrenergic receptors. Neuronal excitation preferentially enhanced noradrenaline-induced synaptic potentiation mediated by D1Rs with minor effects on ß-receptor-dependent potentiation. Increased voluntary exercise by wheel running also enhanced noradrenaline-induced, D1R-mediated synaptic potentiation, suggesting a distinct functional role of the noradrenaline-D1R signaling. We then examined the role of this signaling in antidepressant effects using mice exposed to chronic restraint stress. In the stressed mice, an antidepressant acting on the noradrenergic system induced a mature-to-immature change in the DG neuron phenotype, a previously proposed cellular substrate for antidepressant action. This effect was evident only in mice subjected to wheel running and blocked by a D1R antagonist. These results suggest a critical role of noradrenaline-induced activation of D1Rs in antidepressant effects in DG. Experience-dependent regulation of noradrenaline-D1R signaling may determine responsiveness to antidepressant drugs in depressive disorders.


Subject(s)
Dentate Gyrus , Depressive Disorder , Dopamine , Dopaminergic Neurons , Norepinephrine , Receptors, Dopamine D1 , Animals , Antidepressive Agents/pharmacology , Dentate Gyrus/metabolism , Depressive Disorder/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Mice , Norepinephrine/metabolism , Norepinephrine/pharmacology , Receptors, Dopamine D1/metabolism
8.
Neurosci Res ; 2022 Aug 27.
Article in English | MEDLINE | ID: mdl-36030966

ABSTRACT

The dentate gyrus (DG) of the hippocampus has been implicated in the regulation of stress responses, and in the pathophysiology and treatment of depression. This review discusses the cellular changes caused by chronic stress and the cellular role of the DG in stress-induced behavioral changes and its antidepressant-like effects. Regarding adult-born neurogenic processes in the DG, chronic stress, such as repeated social defeat, suppresses cell proliferation during and immediately after stress; however, this effect is transient. The subsequent differentiation and survival processes are differentially regulated depending on the timing and sensitivity of stress. The activation of young adult-born neurons during stress contributes to stress resilience, while the transient increase in the survival of adult-born neurons after the cessation of stress seems to promote stress susceptibility. In mature granule neurons, the predominant cells in the DG, synaptic plasticity is suppressed by chronic stress. However, a group of mature granule neurons is activated by chronic stress. Chronic antidepressant treatment can transform mature granule neurons to a phenotype resembling that of immature neurons, characterized as "dematuration". Adult-born neurons suppress the activation of mature granule neurons during stress, indicating that local neural interactions within the DG are important for the stress response. Elucidating the stress-associated context- and timing-dependent cellular changes and functions in the DG will provide insights into stress-related psychiatric diseases.

9.
Neurochem Res ; 47(9): 2839-2855, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35907114

ABSTRACT

Astrocytes, together with microglia, play important roles in the non-infectious inflammation and scar formation at the brain infarct during ischemic stroke. After ischemia occurs, these become highly reactive, accumulate at the infarction, and release various inflammatory signaling molecules. The regulation of astrocyte reactivity and function surrounding the infarction largely depends on intercellular communication with microglia. However, the mechanisms involved remain unclear. Furthermore, recent molecular biological studies have revealed that astrocytes are highly divergent under both resting and reactive states, whereas it has not been well reported how the communication between microglia and astrocytes affects astrocyte divergency during ischemic stroke. Minocycline, an antibiotic that reduces microglial activity, has been used to examine the functional roles of microglia in mice. In this study, we used a mouse photothrombotic ischemic stroke model to examine the characteristics of astrocytes after the administration of minocycline during ischemic stroke. Minocycline increased astrocyte reactivity and affected the localization of astrocytes in the penumbra region. Molecular characterization revealed that the induced expression of mRNA encoding the fatty acid binding protein 7 (FABP7) by photothrombosis was enhanced by the minocycline administration. Meanwhile, minocycline did not significantly affect the phenotype or class of astrocytes. The expression of Fabp7 mRNA was well correlated with that of tumor-necrosis factor α (TNFα)-encoding Tnf mRNA, indicating that a correlated expression of FABP7 from astrocytes and TNFα is suppressed by microglial activity.


Subject(s)
Ischemic Stroke , Stroke , Animals , Astrocytes/metabolism , Brain Infarction/metabolism , Disease Models, Animal , Mice , Microglia/metabolism , Minocycline/metabolism , Minocycline/pharmacology , Minocycline/therapeutic use , RNA, Messenger/metabolism , Stroke/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Behav Brain Res ; 416: 113536, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34416303

ABSTRACT

Increasing evidence has shown that adult hippocampal neurogenesis is closely related to the pathophysiological condition of depressive disorders. Recently, chronic social defeat stress paradigms have been regarded as important animal models of depression, accompanied with neural plastic changes in the hippocampus. However, little is known about influences of non-physical stress on neurogenesis. In the present study, we focused on the chronic vicarious social defeat stress paradigm and examined the effect of psychological stress on mouse hippocampal neurogenesis. Immediately after the chronic psychological stress, the cell survival rate in the dentate gyrus of the hippocampus was significantly diminished without modifying the cell proliferation rate. The decreased ratio in cell survival persisted for 4 weeks after the stress-loading period, while the differentiation and maturity of new-born neurons were identical to control groups. Furthermore, treatment with the chronic antidepressant fluoxetine reversed the social behavioral deficits and promoted new-born neurons survival. These results demonstrate that emotional stress in the vicarious social defeat stress paradigm influences neuronal cell survival in the hippocampus, which reinforces its validity as an animal model of depression.


Subject(s)
Antidepressive Agents/pharmacology , Cell Survival/drug effects , Fluoxetine , Hippocampus/drug effects , Neurogenesis , Social Defeat , Animals , Disease Models, Animal , Fluoxetine/antagonists & inhibitors , Fluoxetine/pharmacology , Male , Mice , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/physiology
11.
Neuropsychopharmacol Rep ; 41(1): 56-64, 2021 03.
Article in English | MEDLINE | ID: mdl-33426813

ABSTRACT

AIMS: Electroconvulsive seizure (ECS) therapy is highly effective in the treatment of several psychiatric disorders, including depression. Past studies have shown that the rodent model of ECS reveals the activation of multiple brain regions including the hypothalamus, suggesting that this method of brain stimulation broadly regulates central neuronal function, which results in peripheral function. The ventromedial nucleus of the hypothalamus (VMH) plays an important role in feeding and energy homeostasis. Our previous study showed that ECS increases the expression of anorexigenic factors in the VMH and has an anorexigenic effect in a mouse model. Since the VMH is also suggested to play a critical role in the peripheral lipid metabolism of white adipose tissue (WAT), we hypothesized that ECS alters lipid metabolism via activation of the VMH. METHODS AND RESULTS: Here, we demonstrate that repeated ECS suppresses the fat mass of epididymal WAT and significantly increases the expression levels of lipolytic and brown adipose tissue markers such as Adrb3, Hsl/Lipe, and Ppargc1a. In the VMH, ECS increased the expression of multiple genes, notably Bdnf, Adcyap1, and Crhr2, which are not only anorexigenic factors but are also modulators of lipid metabolism. Furthermore, gold-thioglucose-induced hypothalamic lesions affecting the VMH abolished the effect of ECS on the WAT, indicating that hypothalamus activation is required for the phenotypic changes seen in the epididymal WAT. CONCLUSION: Our data demonstrates a new effect of ECS on the lipid metabolism of WAT via induction of hypothalamic activity involving the VMH.


Subject(s)
Adipose Tissue, White/metabolism , Electroshock , Gene Expression/genetics , Hypothalamus, Middle/metabolism , Lipid Metabolism/physiology , Lipolysis/genetics , Weight Gain/physiology , Animals , Behavior, Animal/physiology , Epididymis/metabolism , Hypothalamus, Middle/pathology , Locomotion/physiology , Male , Mice
12.
J Neurochem ; 157(4): 1196-1206, 2021 05.
Article in English | MEDLINE | ID: mdl-33156548

ABSTRACT

Intracellular signaling pathways that promote axon regeneration are closely linked to the mechanism of neurite outgrowth. TC10, a signaling molecule that acts on neurite outgrowth through membrane transport, is a member of the Rho family G proteins. Axon injury increases the TC10 levels in motor neurons, suggesting that TC10 may be involved in axon regeneration. In this study, we tried to understand the roles of TC10 in the nervous system using TC10 knock-out mice. In cultured hippocampal neurons, TC10 ablation significantly reduced axon elongation without affecting ordinary polarization. We determined a role of TC10 in microtubule stabilization at the growth cone neck; therefore, we assume that TC10 limits axon retraction and promotes in vitro axon outgrowth. In addition, there were no notable differences in the size and structure of brains during prenatal and postnatal development between wild-type and TC10 knock-out mice. In motor neurons, axon regeneration after injury was strongly suppressed in mice lacking TC10 (both in conventional and injured nerve specific deletion). In retinal ganglion cells, TC10 ablation suppressed the axon regeneration stimulated by intraocular inflammation and cAMP after optic nerve crush. These results show that TC10 plays an important role in axon regeneration in both the peripheral and central nervous systems, and the role of TC10 in peripheral axon regeneration is neuron-intrinsic.


Subject(s)
Axons/metabolism , Nerve Regeneration/physiology , rho GTP-Binding Proteins/metabolism , Animals , Hippocampus , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Outgrowth/physiology , Neurons/metabolism , Signal Transduction/physiology
13.
Neurosci Lett ; 739: 135406, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32987131

ABSTRACT

A strong therapeutic target of ischemic stroke is controlling brain inflammation. Recent studies have implicated the critical role of C-C chemokine receptor 5 (CCR5) in neuroinflammation during ischemic stroke. It has been reported that the expression of the matrix metalloproteinases, MMP-3, MMP-12, and MMP-13, is controlled by CCR5; however, their expressional regulation in the infarct brain has not been clearly understood. This study investigated the mRNA expression of Mmp-3, -12, and -13 in the ischemic cerebral cortex of photothrombosis mouse model. The three Mmps were highly upregulated in the early stages of ischemic stroke and were expressed in different types of cells. Mmp-3 and Mmp-13 were expressed in blood vessel endothelial cells after ischemia-induction, whereas Mmp-12 was expressed in activated microglia. The expression of Mmp-13 in resting microglia and in neurons of uninjured cerebral cortex was lost in the infarct region. Therefore, the MMPs responding to CCR5 are differentially regulated during ischemic stroke.


Subject(s)
Cerebral Infarction/metabolism , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Stroke/metabolism , Animals , Disease Models, Animal , Mice, Inbred C57BL , RNA, Messenger , Up-Regulation
14.
iScience ; 23(4): 101025, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32283526

ABSTRACT

The hippocampal mossy fiber (MF) synapse has been implicated in the pathophysiology and treatment of psychiatric disorders. Alterations of dopaminergic and serotonergic modulations at this synapse are candidate mechanisms underlying antidepressant and other related treatments. However, these monoaminergic modulations share the intracellular signaling pathway at the MF synapse, which implies redundancy in their functions. We here show that endogenous monoamines can potentiate MF synaptic transmission in mouse hippocampal slices by activating the serotonin 5-HT4 receptor. Dopamine receptors were not effectively activated by endogenous agonists, suggesting that the dopaminergic modulation is latent. Electroconvulsive treatment enhanced the 5-HT4 receptor-mediated serotonergic synaptic potentiation specifically at the MF synapse, increased the hippocampal serotonin content, and produced an anxiolytic-like behavioral effect in a 5-HT4 receptor-dependent manner. These results suggest that serotonin plays a predominant role in monoaminergic modulations at the MF synapse. Augmentation of this serotonergic modulation may mediate anxiolytic effects of electroconvulsive treatment.

15.
Pharmacol Biochem Behav ; 186: 172767, 2019 11.
Article in English | MEDLINE | ID: mdl-31491434

ABSTRACT

Approximately one third of patients with depression remain treatment resistant with existing antidepressants, suggesting that the currently-available antidepressants cannot induce appropriate responses in the brains of all patients. Long-term exposure to adrenocorticotrophic hormone (ACTH) has been proposed as a model that mimics at least some aspects of clinical treatment-resistant depression in rodents. The purpose of this study was to explore potential causes of antidepressant treatment resistance using the chronic ACTH-treated mouse model. We subjected ACTH-treated mice to a rodent model of electroconvulsive therapy, i.e., electroconvulsive seizure (ECS), which induces various molecular and cellular changes, including in gene expression and adult neurogenesis in the hippocampus. First, behavioral effect of repeated ECS in the forced swim test (FST) was examined. In our experimental setting, ACTH-treated mice showed resistance to the antidepressant-like effect of ECS in the FST. We then examined which cellular and molecular changes induced by ECS were attenuated by ACTH administration. Chronic ACTH treatment suppressed the increase of gene expression such as of Bdnf, Npy, and Drd1 induced by ECS in the hippocampus. In contrast, there was no difference in ECS-induced promotion of the early neurogenetic process in the hippocampus between ACTH-treated and control mice. Our results suggest the possibility that impaired neuromodulation and monoamine signaling in the hippocampus are among the factors contributing to antidepressant treatment resistance.


Subject(s)
Adrenocorticotropic Hormone/administration & dosage , Electroconvulsive Therapy , Adrenocorticotropic Hormone/pharmacology , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/genetics , Male , Mice , Models, Biological , Neurogenesis , Neuropeptide Y/genetics , RNA, Messenger/metabolism , Receptors, Dopamine D1/genetics
16.
J Neurochem ; 149(4): 488-498, 2019 05.
Article in English | MEDLINE | ID: mdl-30825322

ABSTRACT

Electroconvulsive seizure (ECS), a model of electroconvulsive therapy in rodents, strongly increases neurogenesis in the adult hippocampus. Neurogenesis is a multi-step process that spans proliferation, survival, neuronal differentiation, and functional maturation. Our previous study demonstrated that ECS stimulates the proliferation of neural stem-like cells. However, the contribution of ECS to survival, neuronal differentiation, and maturation in newborn cells remains unknown. To evaluate the effect of ECS on these processes, we labeled newborn cells with bromodeoxyuridine (BrdU) before ECS treatment to determine the cell age and examined the survival rate and expression of cellular markers in the BrdU-labeled cells. Our results revealed that exposure to ECS (11 repetitions) during the differentiation phase significantly increased survival and promoted neuronal differentiation of newborn cells in the dentate gyrus. Four of ECS repetitions during the early differentiation phase were sufficient to promote dendritic outgrowth in immature neurons and enhance the expression of the immature neuronal marker, calretinin, in newborn cells. In contrast, exposure to ECS (11 repetitions) during the late maturation phase significantly suppressed the expression of the mature neuronal marker, calbindin, in newborn neurons. These results demonstrate that ECS during the differentiation phase promoted survival and neuronal differentiation and, in contrast, suppressed mature marker expression during the late maturation phase, suggesting that ECS has multiple effects on the different stages of adult neurogenesis.


Subject(s)
Electroshock , Hippocampus/cytology , Neurogenesis/physiology , Neurons/cytology , Aging , Animals , Cell Survival/physiology , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/physiology
17.
Cell Stem Cell ; 24(1): 79-92.e6, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30581080

ABSTRACT

In many tissues, homeostasis is maintained by physical contact between stem cells and an anatomically defined niche. However, how stem cell homeostasis is achieved in environments where cells are motile and dispersed among their progeny remains unknown. Using murine spermatogenesis as a model, we find that spermatogenic stem cell density is tightly regulated by the supply of fibroblast growth factors (FGFs) from lymphatic endothelial cells. We propose that stem cell homeostasis is achieved through competition for a limited supply of FGFs. We show that the quantitative dependence of stem cell density on FGF dosage, the biased localization of stem cells toward FGF sources, and stem cell dynamics during regeneration following injury can all be predicted and explained within the framework of a minimal theoretical model based on "mitogen competition." We propose that this model provides a generic and robust mechanism to support stem cell homeostasis in open, or facultative, niche environments.


Subject(s)
Fibroblast Growth Factor 5/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Homeostasis , Mitogens/pharmacology , Spermatogenesis , Spermatozoa/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Self Renewal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatozoa/physiology , Stem Cells/drug effects , Stem Cells/physiology
18.
Neuron ; 99(3): 464-479.e7, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30033154

ABSTRACT

Repeated environmental stress has been proposed to induce neural inflammation together with depression and anxiety. Innate immune receptors, such as Toll-like receptors (TLRs), are activated by exogenous or endogenous ligands to evoke inflammation. Here we show that the loss of TLR2 and TLR4 (TLR2/4) abolished repeated social defeat stress (R-SDS)-induced social avoidance and anxiety in mice. TLR2/4 deficiency mitigated R-SDS-induced neuronal response attenuation, dendritic atrophy, and microglial activation in the medial prefrontal cortex (mPFC). Furthermore, mPFC microglia-specific TLR2/4 knockdown blocked social avoidance. Transcriptome analyses revealed that R-SDS induced IL-1α and TNF-α in mPFC microglia in a TLR2/4-dependent manner, and antibody blockade of these cytokines in the mPFC suppressed R-SDS-induced social avoidance. These results identify TLR2/4 as crucial mediators of R-SDS-induced microglial activation in the mPFC, which leads to neuronal and behavioral changes through inflammation-related cytokines, highlighting unexpected pivotal roles of innate immunity in the mPFC in repeated environmental stress-induced behavioral changes. VIDEO ABSTRACT.


Subject(s)
Avoidance Learning/physiology , Microglia/metabolism , Prefrontal Cortex/metabolism , Stress, Psychological/metabolism , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 4/deficiency , Animals , Cells, Cultured , HEK293 Cells , Humans , Immunity, Innate/physiology , Interpersonal Relations , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/transplantation , Prefrontal Cortex/cytology , Prefrontal Cortex/immunology , Stress, Psychological/immunology , Stress, Psychological/psychology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology
19.
Neuroreport ; 29(3): 174-180, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29215465

ABSTRACT

Middle cerebral artery occlusion (MCAO) is the most widely used animal model of ischemic stroke. This model well recapitulates the pathological features of most human cases; however, MCAO is technically difficult to achieve in mice and has some disadvantages for investigating the molecular mechanisms of pathological progression in stroke. The recently developed photothrombosis model may be more suitable for research on the molecular mechanisms of ischemic stroke in mice. Yet, similarities and differences between the photothrombosis and MCAO models are not well characterized. In the present study, we examined the expression of tissue inhibitor of metalloproteinases (TIMPs) and matrix metalloproteinases (MMPs) in the brains of photothrombosis model mice. Our results indicated that the gene expression of TIMP-1 was upregulated in endothelial cells in the pathological area surrounding the infarction, similar to the MCAO model. Yet, pathologically induced changes in TIMP-1 were not affected by treatment with aspirin or etodolac. Whereas MMP-2 and MMP-8 mRNA were upregulated after infarction in both models, MMP-9 expression, which is induced in the infarct area in the MCAO model, was unchanged in the photothrombosis model. These findings suggest that the expression patterns of TIMP-1 and MMP-9 are regulated independently in photothrombosis model mice.


Subject(s)
Brain/metabolism , Ischemia/metabolism , Matrix Metalloproteinases/metabolism , Stroke/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Brain/drug effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Etodolac/pharmacology , Gene Expression/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Ischemia/drug therapy , Male , Mice, Inbred C57BL , RNA, Messenger/metabolism , Stroke/drug therapy , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL