Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Pharmacol Sci ; 154(4): 279-293, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38485346

ABSTRACT

Despite the importance of lipid mediators in stress and depression and their link to inflammation, the influence of stress on these mediators and their role in inflammation is not fully understood. This study used RNA-seq, LC-MS/MS, and flow cytometry analyses in a mouse model subjected to chronic social defeat stress to explore the effects of acute and chronic stress on lipid mediators, gene expression, and cell population in the bone marrow and spleen. In the bone marrow, chronic stress induced a sustained transition from lymphoid to myeloid cells, accompanied by corresponding changes in gene expression. This change was associated with decreased levels of 15-deoxy-d12,14-prostaglandin J2, a lipid mediator that inhibits inflammation. In the spleen, chronic stress also induced a lymphoid-to-myeloid transition, albeit transiently, alongside gene expression changes indicative of extramedullary hematopoiesis. These changes were linked to lower levels of 12-HEPE and resolvins, both critical for inhibiting and resolving inflammation. Our findings highlight the significant role of anti-inflammatory and pro-resolving lipid mediators in the immune responses induced by chronic stress in the bone marrow and spleen. This study paves the way for understanding how these lipid mediators contribute to the immune mechanisms of stress and depression.


Subject(s)
Bone Marrow , Spleen , Mice , Animals , Spleen/metabolism , Bone Marrow/metabolism , Chromatography, Liquid , Tandem Mass Spectrometry , Inflammation/metabolism , Lipids , Gene Expression
2.
Brain Behav Immun ; 116: 329-348, 2024 02.
Article in English | MEDLINE | ID: mdl-38142917

ABSTRACT

BACKGROUND: Latent chronic inflammation has been proposed as a key mediator of multiple derangements in metabolic syndrome (MetS), which are increasingly becoming recognized as risk factors for age-related cognitive decline. However, the question remains whether latent chronic inflammation indeed induces brain inflammation and cognitive decline. METHODS: A mouse model of latent chronic inflammation was constructed by a chronic subcutaneous infusion of low dose lipopolysaccharide (LPS) for four weeks. A receptor for advanced glycation end products (RAGE) knockout mouse, a chimeric myeloid cell specific RAGE-deficient mouse established by bone marrow transplantation and a human endogenous secretory RAGE (esRAGE) overexpressing adenovirus system were utilized to examine the role of RAGE in vivo. The cognitive function was examined by a Y-maze test, and the expression level of genes was determined by quantitative RT-PCR, western blot, immunohistochemical staining, or ELISA assays. RESULTS: Latent chronic inflammation induced MetS features in C57BL/6J mice, which were associated with cognitive decline and brain inflammation characterized by microgliosis, monocyte infiltration and endothelial inflammation, without significant changes in circulating cytokines including TNF-α and IL-1ß. These changes as well as cognitive impairment were rescued in RAGE knockout mice or chimeric mice lacking RAGE in bone marrow cells. P-selectin glycoprotein ligand-1 (PSGL-1), a critical adhesion molecule, was induced in circulating mononuclear cells in latent chronic inflammation in wild-type but not RAGE knockout mice. These inflammatory changes and cognitive decline induced in the wild-type mice were ameliorated by an adenoviral increase in circulating esRAGE. Meanwhile, chimeric RAGE knockout mice possessing RAGE in myeloid cells were still resistant to cognitive decline and brain inflammation. CONCLUSIONS: These findings indicate that RAGE in inflammatory cells is necessary to mediate stimuli of latent chronic inflammation that cause brain inflammation and cognitive decline, potentially by orchestrating monocyte activation via regulation of PSGL-1 expression. Our results also suggest esRAGE-mediated inflammatory regulation as a potential therapeutic option for cognitive dysfunction in MetS with latent chronic inflammation.


Subject(s)
Cognitive Dysfunction , Encephalitis , Metabolic Syndrome , Animals , Humans , Mice , Inflammation , Mice, Inbred C57BL , Mice, Knockout , Receptor for Advanced Glycation End Products
3.
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.

4.
Cells ; 12(13)2023 07 05.
Article in English | MEDLINE | ID: mdl-37443823

ABSTRACT

Inflammation has been associated with depression, and innate immune receptors, such as the Toll-like receptor (TLR) 2/4 in the medial prefrontal cortex (mPFC), are crucial for chronic stress-induced depression-related behaviors in mice. HMGB1, a putative ligand for TLR2/4, has been suggested to promote depression-related behaviors under acute stress. However, the roles of endogenous HMGB1 under chronic stress remain to be investigated. Here, we found that the cerebroventricular infusion of HMGB1 proteins blocked stress-induced social avoidance and that HMGB1-neutralizing antibodies augmented repeated social defeat stress-induced social avoidance in mice, suggesting the antidepressive-like effect of HMGB1 in the brain. By contrast, the infusion of HMGB1-neutralizing antibodies to the mPFC and HMGB1 knockout in α-CaMKII-positive forebrain neurons attenuated the social avoidance, suggesting the pro-depressive-like effect of HMGB1 released from prefrontal neurons under chronic stress. In addition, repeated social defeat stress induced HMGB1 nuclear export selectively in mPFC neurons, which was abolished in the mice lacking RAGE, one of HMGB1 receptors, suggesting the positive feedback loop of HMGB1-RAGE signaling under chronic stress. These findings pave the way for identifying multiple roles of HMGB1 in the brain for chronic stress and depression.


Subject(s)
HMGB1 Protein , Social Defeat , Animals , Mice , Active Transport, Cell Nucleus , Social Behavior , Neurons
5.
J Biochem ; 173(3): 153-157, 2023 Mar 07.
Article in English | MEDLINE | ID: mdl-36539335

ABSTRACT

Microglia are immune cells in the central nervous system that engulf unnecessary synapses during development. In vivo imaging has substantially improved in recent years, besides the development of tools for manipulating microglia and neurons. These techniques reveal the novel functions of microglia. Microglia regulate neuronal activity to prevent synchronization. This neuron-microglia interaction is mediated by adenosine triphosphate-P2Y12 and adenosine-adenosine A1 receptor signalling in the striatum. Moreover, microglia release inflammation-related molecules that suppress neuronal activity, thus leading to lipopolysaccharide-induced aversion. Prostaglandin E2 (PGE2)-PGE receptor 1 signalling in the striatum underlies this behavioural alteration. Chronic stress activates microglia through toll-like receptor (TLR) 2 and TLR4 to release pro-inflammatory cytokines in the medial prefrontal cortex, thereby causing social avoidance. Microglia play multiple functions under physiological conditions, as well as pathological and psychological stress.


Subject(s)
Microglia , Neurons , Humans , Microglia/pathology , Inflammation/pathology , Cytokines , Signal Transduction
6.
Sci Rep ; 12(1): 11385, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35790870

ABSTRACT

Severe and prolonged social stress induces mood and cognitive dysfunctions and precipitates major depression. Neuroinflammation has been associated with chronic stress and depression. Rodent studies showed crucial roles of a few inflammation-related lipid mediators for chronic stress-induced depressive-like behaviors. Despite an increasing number of lipid mediators identified, systematic analyses of synthetic pathways of lipid mediators in chronic stress models have not been performed. Using LC-MS/MS, here we examined the effects of chronic social defeat stress on multiple synthetic pathways of lipid mediators in brain regions associated with stress susceptibility in mice. Chronic social defeat stress increased the amounts of 12-lipoxygenase (LOX) metabolites, 12-HETE and 12-HEPE, specifically in the nucleus accumbens 1 week, but not immediately, after the last stress exposure. The increase was larger in stress-resilient mice than stress-susceptible mice. The S isomer of 12-HETE was selectively increased in amount, indicating the role of 12S-LOX activity. Among the enzymes known to have 12S-LOX activity, only Alox12 mRNA was reliably detected in the brain and enriched in brain endothelial cells. These findings suggest that chronic social stress induces a late increase in the amounts of 12S-LOX metabolites derived from the brain vasculature in the nucleus accumbens in a manner associated with stress resilience.


Subject(s)
Nucleus Accumbens , Social Defeat , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Animals , Arachidonate 12-Lipoxygenase/metabolism , Chromatography, Liquid , Endothelial Cells/metabolism , Mice , Mice, Inbred C57BL , Nucleus Accumbens/metabolism , Tandem Mass Spectrometry
7.
J Pharmacol Sci ; 149(3): 166-171, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35641029

ABSTRACT

Microglia have diverse physiological and pathological functions. However, the transcriptional mechanisms remain elusive. Here we sought new transcription factors relevant to microglial functions from the microglial transcriptome of stressed mice and evaluated their roles in primary microglia. TLR2 and TLR4 agonists increased Rel, Atf3, and Cebpb and decreased Hhex in primary microglia as repeated social defeat stress. Although Hhex was not studied in microglia, TLR2 and TLR4 agonists decreased Hhex, and Hhex overexpression attenuated TLR4-increased expression of inflammation-related genes. These findings suggest that Hhex negatively regulates inflammation-related genes in microglia and that TLR2/4 activation reduces Hhex, facilitating TLR4-mediated neuroinflammation.


Subject(s)
Homeodomain Proteins , Microglia , Transcription Factors , Animals , Homeodomain Proteins/metabolism , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Toll-Like Receptor 2/agonists , Toll-Like Receptor 4/agonists , Transcription Factors/genetics , Transcription Factors/metabolism
8.
J Pharmacol Sci ; 148(2): 262-266, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35063142

ABSTRACT

Currently used antidepressant drugs target and facilitate the action of monoamine neurotransmission. However, approximately 30% of patients do not respond to these drugs. Therefore, there is an urgent need to develop novel therapeutic targets. Several clinical studies have reported that inflammatory cytokines and neutrophils are increased in the blood of patients with major depression. Since social and environmental stress is a risk factor for mental illnesses such as major depression, many research groups have employed chronic stress models in which mice are repeatedly exposed to stressful events. Chronic stress induces neuroinflammation originating from microglia in the medial prefrontal cortex, leading to depressive-like behavior. Moreover, chronic stress influences peripheral immune cells by activating the sympathetic nervous system and the hypothalamus-pituitary-adrenal gland axis. The infiltration of monocytes expressing interleukin (IL)-1ß into the brain is involved in chronic stress-induced elevated anxiety. The penetration of IL-6 derived from monocytes into the nucleus accumbens is involved in chronic stress-induced depression-like behavior. Furthermore, cell-cell and peripheral brain interactions and their molecular basis have been discovered. These findings may pave the way for the development of biological markers and therapeutic drugs.


Subject(s)
Behavior/physiology , Brain/pathology , Depression/etiology , Depression/pathology , Inflammation Mediators/metabolism , Inflammation/pathology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Animals , Brain/metabolism , Chronic Disease , Depression/psychology , Depression/therapy , Disease Models, Animal , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mice , Molecular Targeted Therapy , Stress, Psychological/complications
9.
Br J Pharmacol ; 178(4): 827-844, 2021 02.
Article in English | MEDLINE | ID: mdl-32678951

ABSTRACT

BACKGROUND AND PURPOSE: Inflammation has been associated with stress-related mental disturbances. Rodent studies have reported that blood-borne cytokines are crucial for stress-induced changes in emotional behaviours. However, the roles and regulation of leukocytes in chronic stress remain unclear. EXPERIMENTAL APPROACH: Adult male C57BL/6N mice were subjected to repeated social defeat stress (R-SDS) with two protocols which differed in stress durations, stress cycles, and housing conditions, followed by the social interaction test. The numbers of leukocyte subsets in the bone marrow, spleen, and blood were determined by flow cytometry shortly after or several days after R-SDS. These leukocyte changes were studied in two strains of mice with different stress susceptibility, C57BL/6N and BALB/c mice. KEY RESULTS: R-SDS with both protocols similarly induced social avoidance in C57BL/6N mice. In the bone marrow, neutrophils and monocytes were increased, and T cells, B cells, NK cells, and dendritic cells were decreased with both protocols. In the blood, neutrophils and monocytes were increased with both protocols, whereas T cells, B cells, NK cells, and dendritic cells were decreased with one of these. Neutrophils and monocytes were also increased in the spleen. Changes in the bone marrow and increased levels of circulating neutrophils were maintained for 6 days after R-SDS. BALB/c mice showed greater social avoidance and increase in circulating neutrophils than C57BL/6N mice. CONCLUSION AND IMPLICATIONS: In two strains of mice, chronic stress induced neutrophil mobilization and its maintenance. These effects were strain-related and may contribute to the pathology of mental illness. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.4/issuetoc.


Subject(s)
Neutrophils , Social Defeat , Animals , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Stress, Psychological
10.
Int J Mol Sci ; 21(24)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348553

ABSTRACT

As daily lifestyle is closely associated with mental illnesses, diet-based preventive approaches are receiving attention. Supplementation with hop bitter acids such as iso-α-acids (IAA) and mature hop bitter acids (MHBA) improves mood states in healthy older adults. However, the underlying mechanism remains unknown. Since acute oral consumption with IAA increases dopamine levels in hippocampus and improves memory impairment via vagal nerve activation, here we investigated the effects of chronic administration of hop bitter acids on the dopaminergic activity associated with emotional disturbance in a mouse model of repeated social defeat stress (R-SDS). Chronic administration of IAA and MHBA significantly increased dopaminergic activity based on the dopamine metabolite to dopamine ratio in the hippocampus and medial prefrontal cortex following R-SDS. Hippocampal dopaminergic activity was inversely correlated with the level of R-SDS-induced social avoidance with or without IAA administration. Therefore, chronic treatment with hop bitter acids enhances stress resilience-related hippocampal dopaminergic activity.


Subject(s)
Cyclohexenes/administration & dosage , Dopamine/metabolism , Hippocampus/metabolism , Humulus/chemistry , Plant Extracts/administration & dosage , Social Defeat , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Terpenes/administration & dosage , Affective Symptoms/drug therapy , Animals , Behavior, Animal/drug effects , Cyclohexenes/chemistry , Disease Models, Animal , Isomerism , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Plant Extracts/chemistry , Social Interaction/drug effects , Terpenes/chemistry
11.
Nihon Yakurigaku Zasshi ; 155(6): 390-394, 2020.
Article in Japanese | MEDLINE | ID: mdl-33132256

ABSTRACT

The current therapeutic drugs for major depression mainly modulate monoaminergic signaling. Since they are not effective for all patients, the development of novel therapeutic target is required. Recently, it has been reported that inflammation-related molecules are increased in the blood from patients with major depression. Therefore, neuroinflammation is a possible cause of these disorders. However, we still do not know whether neuroinflammation induces depression. Since social and environmental stress is a risk factor for mental illnesses, repeated social defeat stress is employed as an animal model of depression. We found that prostaglandin E2 (PGE2) suppresses mesocortical dopaminergic pathway to induce behavioral changes and cyclooxygenase-1 (COX-1), a key enzyme for PGE2 production, is essential for repeated stress-induced PGE2 production and behavioral changes. Based on the finding that COX-1 is expressed in microglia in the brain, we are wondering if microglia plays an important role in stress-induced behavioral changes. We revealed that Toll-like receptor (TLR) 2 and 4 in prefrontal microglia are crucial for repeated stress-induced behavioral changes. Our results indicate that repeated social defeat stress induces microglial activation through TLR2 and 4, thereby leading to neuronal and behavioral changes through proinflammatory cytokines such as TNFα and IL-1α. These findings revealed the essential role and molecular basis of neuroinflammation. In addition, we developed the drug screening platform which targets neuroinflammation for neurodegenerative disease such as amyotrophic lateral sclerosis. Our findings pave the way for the development of therapeutic drugs for major depression targeting neuroinflammation which causes neurological disorders.


Subject(s)
Depression , Drug Evaluation, Preclinical , Inflammation , Neurodegenerative Diseases , Animals , Depression/drug therapy , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Microglia , Stress, Psychological
12.
Nihon Yakurigaku Zasshi ; 155(6): 435, 2020.
Article in Japanese | MEDLINE | ID: mdl-33132263

Subject(s)
Iron , Homeostasis
14.
Sci Rep ; 9(1): 17548, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31772309

ABSTRACT

Inflammation in the brain and periphery has been associated with stress-related pathology of mental illness. We have shown that prostaglandin (PG) E2, an arachidonic acid-derived lipid mediator, and innate immune receptors Toll-like receptor (TLR) 2/4 are crucial for repeated stress-induced behavioral changes in rodents. However, how the stress induces PGE2 synthesis in the brain and whether TLR2/4 are involved in the PGE2 synthesis remain unknown. Using mice lacking TLR2 and TLR4 in combination, here we show that social defeat stress (SDS) induced the PGE2 synthesis in subcortical, but not cortical, tissues in a TLR2/4-dependent manner. It is known that PGE2 in the brain is mainly derived by monoacylglycerol lipase (MAGL)-mediated conversion of endocannabinoid 2-arachidonoylglycerol to free-arachidonic acid, a substrate for cyclooxygenase (COX) for PGE2 synthesis. We found that TLR2/4 deletion reduced the mRNA expression of MAGL and COX1 in subcortical tissues after repeated SDS. Perturbation of MAGL and COX1 as well as COX2 abolished SDS-induced PGE2 synthesis in subcortical tissues. Furthermore, systemic administration of JZL184, an MAGL inhibitor, abolished repeated SDS-induced social avoidance. These results suggest that SDS induces PGE2 synthesis in subcortical regions of the brain via the MAGL-COX pathway in a TLR2/4-dependent manner, thereby leading to social avoidance.


Subject(s)
Brain/metabolism , Dinoprostone/metabolism , Monoacylglycerol Lipases/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Stress, Psychological/metabolism , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , Aggression/physiology , Animals , Brain/physiopathology , Dinoprostone/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Monoacylglycerol Lipases/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Stress, Psychological/enzymology , Stress, Psychological/physiopathology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
15.
Nutrients ; 11(10)2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31618902

ABSTRACT

SCOPE: Peptides containing tryptophan-tyrosine sequences, including the lacto-tetrapeptide glycine-threonine-tryptophan-tyrosine (GTWY) and ß-lactolin, from ß-lactoglobulin in whey enzymatic digestion, enhance hippocampus-dependent memory functions, which are blocked by the systemic administration of dopamine D1-like antagonist. In this study, we investigated the role of the hippocampal dopaminergic system in the memory-enhancing effect of ß-lactolin. METHODS AND RESULTS: The results of in vivo microdialysis revealed that oral administration of ß-lactolin increased the extracellular concentration of dopamine in the hippocampus and enhanced both spatial working memory, as measured in the Y-maze test, and spatial reference memory, as measured in the novel object location test. These memory-enhancing effects of ß-lactolin, but not the baseline memory functions, were impaired by the knockdown of the dopamine D1 receptor subtype in the hippocampus. ß-Lactolin also enhanced object memory, as measured by the novel object recognition test. However, D1 knockdown in the hippocampus spared this memory function either with or without the administration of ß-lactolin. CONCLUSIONS: The present results indicate that oral administration of ß-lactolin increases dopamine release and D1 receptor signaling in the hippocampus, thereby enhancing spatial memory, but it may improve object memory via a separate mechanism.


Subject(s)
Behavior, Animal/drug effects , Dopamine Agonists/pharmacology , Dopamine/metabolism , Hippocampus/drug effects , Nootropic Agents/pharmacology , Oligopeptides/pharmacology , Receptors, Dopamine D1/agonists , Spatial Memory/drug effects , Animals , Hippocampus/metabolism , Male , Maze Learning/drug effects , Memory, Short-Term/drug effects , Mice, Inbred ICR , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Recognition, Psychology/drug effects , Signal Transduction
16.
Nutrients ; 11(9)2019 Sep 09.
Article in English | MEDLINE | ID: mdl-31505850

ABSTRACT

The number of patients with mental illnesses is rapidly increasing, and daily lifestyle is closely associated with the development of symptoms. It is suggested that inflammatory molecules derived from microglia play crucial roles for the pathophysiology of depression. In the present study, we discovered that leucine-histidine (LH) dipeptide suppressed activation of primary microglia. The effects of LH dipeptide orally administered were measured using tail suspension test (TST) in mice injected with lipopolysaccharide and social interaction test in mice received social defeat stress. LH dipeptide reduced pro-inflammatory cytokines upon stimulation in microglia. Orally administered LH dipeptide was delivered to the brain and suppressed the production of pro-inflammatory cytokines in the brain and concomitant depression-like behavior in the TST. Moreover, oral administration of LH dipeptide suppressed the induction of depression- and anxiety-like behaviors induced by repeated social defeat stress. These results indicate that LH dipeptide suppressed the activation of microglia and ameliorated depression-associated emotional disturbances. Further, we found that LH dipeptide was abundant in various fermented products. Together with previous epidemiological reports that daily intake of these fermented foods is negatively associated with the incidence of psychiatric diseases, our findings suggest that food rich in LH dipeptide may improve mental health.


Subject(s)
Affective Symptoms/drug therapy , Dipeptides/pharmacology , Encephalitis/drug therapy , Microglia/drug effects , Stress, Psychological/drug therapy , Affective Symptoms/physiopathology , Affective Symptoms/psychology , Animals , Anxiety/psychology , Behavior, Animal/drug effects , Cytokines/drug effects , Depression/psychology , Encephalitis/physiopathology , Encephalitis/psychology , Mice , Social Behavior , Stress, Psychological/physiopathology , Stress, Psychological/psychology
17.
Mol Brain ; 12(1): 68, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31370877

ABSTRACT

Post-traumatic stress disorder (PTSD) is a psychiatric disorder associated with memories of traumatic experiences. Recent studies have shown that the forgetting of contextual fear memory is promoted via increased adult hippocampal neurogenesis induced by neurogenesis enhancers, such as memantine (MEM) and exercise, raising the possibility that neurogenesis enhancers improve PTSD by facilitating the forgetting of traumatic memory. On the other hand, repeated exposure to social defeat (SD) stress by aggressor mice induces social avoidance behavior to the aggressor and chronic anxiety-like behavior. In this study, we assumed this SD stress paradigm as a PTSD-like model and examined the effects of treatment with neurogenesis enhancer MEM on SD stress-induced PTSD-like behavior. Male C57BL/6 mice received SD stress for 10 consecutive days and were assessed for social avoidance memory to the aggressor (memory of aggressor mice) and anxiety-like behavior using social interaction and elevated zero maze tasks. Consistent with previous studies, SD mice formed social avoidance memory and exhibited increased anxiety-like behavior. Importantly, subsequent MEM treatment (once a week for 4 weeks) significantly reduced social avoidance behavior, suggesting that MEM-treated SD mice showed forgetting of social avoidance memory. Interestingly, MEM-treated SD mice showed comparable anxiety-like behavior with control mice that were not exposed to SD stress. Moreover, MEM-treated SD mice showed no reinstatement of social avoidance memory following single re-exposure to the aggressor. Our findings suggest that neurogenesis enhancer not only enhanced the forgetting of traumatic memory but also improved PTSD (anxiety)-like behavior.


Subject(s)
Behavior, Animal , Hippocampus/physiopathology , Memantine/pharmacology , Memory , Neurogenesis/drug effects , Social Behavior , Stress Disorders, Post-Traumatic/complications , Stress, Psychological/complications , Animals , Anxiety/complications , Anxiety/physiopathology , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Hippocampus/drug effects , Male , Memory/drug effects , Mice, Inbred C57BL , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/physiopathology
18.
Psychiatry Clin Neurosci ; 73(11): 669-675, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31215710

ABSTRACT

Stress caused by adverse and demanding conditions, a risk factor for mental illnesses, induces adaptive or maladaptive neural and behavioral consequences, depending on the conditions. Studies using rodent stress models have revealed multiple mechanisms related to dopamine and inflammation for stress-induced neural and behavioral changes. Thus, repeated stress alters activities of ventral tegmental area dopamine neurons projecting to the nucleus accumbens and the medial prefrontal cortex in distinct manners. In the nucleus accumbens, repeated stress decreases activities of D1 receptor-expressing neurons. In the medial prefrontal cortex, single stress increases dopamine D1 receptor signaling, leading to dendritic hypertrophy of excitatory neurons and stress resilience. These changes are attenuated with repetition of stress via prostaglandin E2 , an inflammation-related lipid mediator. Repeated stress activates microglia in the medial prefrontal cortex and the hippocampus. Innate immune receptors, such as the toll-like receptor 2/4 and P2X7, are crucial for repeated stress-induced microglial activation, leading to neural and behavioral changes through proinflammatory cytokines. In addition, repeated stress induces monocyte infiltration to the brain, and impairs the blood-brain barrier in the nucleus accumbens, leading to cytokine leakage to the brain. These monocyte-derived responses are involved in stress-induced behavioral changes. These findings show crucial roles of the accumbal and prefrontal dopamine pathways and inflammatory responses in the brain and body to direct adaptive and maladaptive consequences of stress, and pave the way for identifying a neural origin of stress and understanding the stress-related pathology of mental illnesses.


Subject(s)
Adaptation, Psychological , Dopamine , Inflammation/pathology , Stress, Psychological/pathology , Stress, Psychological/psychology , Animals , Humans , Receptors, Dopamine
19.
Nutrients ; 11(2)2019 Feb 06.
Article in English | MEDLINE | ID: mdl-30736353

ABSTRACT

Tryptophan-tyrosine (WY)-related peptides including the ß-lactopeptide of the glycine-threonine-tryptophan-tyrosine peptide, ß-lactolin, improve spatial memory. However, whether and how the WY dipeptide as the core sequence in WY-related peptides improves memory functions has not been investigated. This study assessed the pharmacological effects of the WY dipeptide on memory impairment to elucidate the mechanisms. Here, we showed that oral administration of dipeptides of WY, tryptophan-methionine (WM), tryptophan-valine, tryptophan-leucine, and tryptophan-phenylalanine improved spontaneous alternation of the Y-maze test in scopolamine-induced amnesic mice. In contrast, tyrosine-tryptophan, methionine-tryptophan, tryptophan, tyrosine, and methionine had no effect. These results indicated that the conformation of dipeptides with N-terminal tryptophan is required for their memory improving effects. WY dipeptide inhibited the monoamine oxidase B activity in vitro and increased dopamine levels in the hippocampus and frontal cortex, whereas tryptophan did not cause these effects. In addition, the treatment with SCH-23390, a dopamine D1-like receptor antagonist, and the knockdown of the hippocampal dopamine D1 receptor partially attenuated the memory improvement induced by the WY dipeptide. Importantly, WY dipeptide improved the spontaneous alternations of the Y-maze test in aged mice. These results suggest that the WY dipeptide restores memory impairments by augmenting dopaminergic activity. The development of supplements rich in these peptides might help to prevent age-related cognitive decline.


Subject(s)
Amnesia/drug therapy , Dipeptides/pharmacology , Dopamine/metabolism , Memory/drug effects , Tryptophan/pharmacology , Tyrosine/pharmacology , Amnesia/chemically induced , Animals , Disease Models, Animal , Frontal Lobe/metabolism , Hippocampus/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Scopolamine
20.
Int Immunol ; 31(9): 579-587, 2019 08 23.
Article in English | MEDLINE | ID: mdl-30810163

ABSTRACT

Prolonged or excessive stress may induce emotional and cognitive disturbances, and is a risk factor for mental illnesses. Using rodent chronic stress models of depression, roles of multiple lipid mediators related to inflammation have been revealed in chronic stress-induced emotional alterations. Prostaglandin (PG) E2, an arachidonic acid (AA)-derived lipid mediator, and its receptor subtype EP1 mediate depression-like behavior induced by repeated social defeat stress through attenuating prefrontal dopaminergic activity. Repeated social defeat stress activates microglia through innate immune receptors, and induces PGE2 synthesis through cyclooxygenase-1, a prostaglandin synthase enriched in microglia. PGD2, another AA-derived lipid mediator, has been implicated in depression induced by chronic stress, although either pro-depressive or anti-depressive actions have been reported. Chronic stress up-regulates hippocampal expression of 5-lipoxygenase, hence synthesis of cysteinyl leukotrienes, thereby inducing depression through their receptors. Consistent with beneficial effects of n-3 fatty acids in the diet of depressive patients, resolvins-a novel class of pro-resolving lipid mediators-in the brain attenuate neuroinflammation-associated depression. These findings in animal models of depression offer lipid mediators and related molecules as novel therapeutic targets for treating depression. To translate these findings into clinics, translational biomarkers to visualize lipid mediator profiles in depressive patients need to be established.


Subject(s)
Depression/drug therapy , Depression/metabolism , Dinoprostone/metabolism , Dinoprostone/therapeutic use , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...