Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.509
Filter
1.
Behav Brain Funct ; 20(1): 18, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965529

ABSTRACT

BACKGROUND: Anxiety disorders are one of the most common mental disorders. Ghrelin is a critical orexigenic brain-gut peptide that regulates food intake and metabolism. Recently, the ghrelin system has attracted more attention for its crucial roles in psychiatric disorders, including depression and anxiety. However, the underlying neural mechanisms involved have not been fully investigated. METHODS: In the present study, the effect and underlying mechanism of ghrelin signaling in the nucleus accumbens (NAc) core on anxiety-like behaviors were examined in normal and acute stress rats, by using immunofluorescence, qRT-PCR, neuropharmacology, molecular manipulation and behavioral tests. RESULTS: We reported that injection of ghrelin into the NAc core caused significant anxiolytic effects. Ghrelin receptor growth hormone secretagogue receptor (GHSR) is highly localized and expressed in the NAc core neurons. Antagonism of GHSR blocked the ghrelin-induced anxiolytic effects. Moreover, molecular knockdown of GHSR induced anxiogenic effects. Furthermore, injection of ghrelin or overexpression of GHSR in the NAc core reduced acute restraint stress-induced anxiogenic effects. CONCLUSIONS: This study demonstrates that ghrelin and its receptor GHSR in the NAc core are actively involved in modulating anxiety induced by acute stress, and raises an opportunity to treat anxiety disorders by targeting ghrelin signaling system.


Subject(s)
Anxiety , Ghrelin , Nucleus Accumbens , Rats, Sprague-Dawley , Receptors, Ghrelin , Signal Transduction , Stress, Psychological , Animals , Ghrelin/metabolism , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Male , Anxiety/metabolism , Anxiety/psychology , Receptors, Ghrelin/metabolism , Receptors, Ghrelin/genetics , Rats , Stress, Psychological/metabolism , Stress, Psychological/psychology , Signal Transduction/drug effects , Signal Transduction/physiology , Behavior, Animal/drug effects
2.
J Neuroinflammation ; 21(1): 166, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956653

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) are mutual risk factors, with both conditions inducing cognitive impairment and anxiety. However, whether OSA exacerbates cognitive impairment and anxiety in patients with T2DM remains unclear. Moreover, TREM2 upregulation has been suggested to play a protective role in attenuating microglia activation and improving synaptic function in T2DM mice. The aim of this study was to explore the regulatory mechanisms of TREM2 and the cognitive and anxiety-like behavioral changes in mice with OSA combined with T2DM. METHODS: A T2DM with OSA model was developed by treating mice with a 60% kcal high-fat diet (HFD) combined with intermittent hypoxia (IH). Spatial learning memory capacity and anxiety in mice were investigated. Neuronal damage in the brain was determined by the quantity of synapses density, the number and morphology of brain microglia, and pro-inflammatory factors. For mechanism exploration, an in vitro model of T2DM combined with OSA was generated by co-treating microglia with high glucose (HG) and IH. Regulation of TREM2 on IFNAR1-STAT1 pathway was determined by RNA sequencing and qRT-PCR. RESULTS: Our results showed that HFD mice exhibited significant cognitive dysfunction and anxiety-like behavior, accompanied by significant synaptic loss. Furthermore, significant activation of brain microglia and enhanced microglial phagocytosis of synapses were observed. Moreover, IH was found to significantly aggravate anxiety in the HFD mice. The mechanism of HG treatment may potentially involve the promotion of TREM2 upregulation, which in turn attenuates the proinflammatory microglia by inhibiting the IFNAR1-STAT1 pathway. Conversely, a significant reduction in TREM2 in IH-co-treated HFD mice and HG-treated microglia resulted in the further activation of the IFNAR1-STAT1 pathway and consequently increased proinflammatory microglial activation. CONCLUSIONS: HFD upregulated the IFNAR1-STAT1 pathway and induced proinflammatory microglia, leading to synaptic damage and causing anxiety and cognitive deficits. The upregulated TREM2 inT2DM mice brain exerted a negative regulation of the IFNAR1-STAT1 pathway. Mice with T2DM combined with OSA exacerbated anxiety via the downregulation of TREM2, causing heightened IFNAR1-STAT1 pathway activation and consequently increasing proinflammatory microglia.


Subject(s)
Anxiety , Diabetes Mellitus, Type 2 , Diet, High-Fat , Hypoxia , Membrane Glycoproteins , Mice, Inbred C57BL , Receptor, Interferon alpha-beta , Receptors, Immunologic , Signal Transduction , Animals , Mice , Diet, High-Fat/adverse effects , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Anxiety/etiology , Anxiety/metabolism , Signal Transduction/physiology , Signal Transduction/drug effects , Hypoxia/metabolism , Hypoxia/complications , Male , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/psychology , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Microglia/metabolism , STAT1 Transcription Factor/metabolism , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/metabolism , Sleep Apnea, Obstructive/psychology
3.
Dev Psychobiol ; 66(6): e22523, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38970242

ABSTRACT

The current literature suggests that relaxin-3/relaxin/insulin-like family peptide receptor 3 (RLN-3/RXFP-3) system is involved in the pathophysiology of affective disorders because the results of anatomical and pharmacological studies have shown that the RLN-3 signaling pathway plays a role in modulating the stress response, anxiety, arousal, depression-like behavior, and neuroendocrine homeostasis. The risk of developing mental illnesses in adulthood is increased by exposure to stress in early periods of life. The available data indicate that puberty is especially characterized by the development of the neural system and emotionality and is a "stress-sensitive" period. The presented study assessed the short-term changes in the expression of RLN-3 and RXFP-3 mRNA in the stress-dependent brain regions in male pubertal Wistar rats that had been subjected to acute stress. Three stressors were applied from 42 to 44 postnatal days (first day: a single forced swim; second day: stress on an elevated platform that was repeated three times; third day: restraint stress three times). Anxiety (open field, elevated plus maze test) and anhedonic-like behavior (sucrose preference test) were estimated during these tests. The corticosterone (CORT) levels and blood morphology were estimated. We found that the RXFP-3 mRNA expression decreased in the brainstem, whereas it increased in the hypothalamus 72 h after acute stress. These molecular changes were accompanied by the increased levels of CORT and anxiety-like behavior detected in the open field test that had been conducted earlier, that is, 24 h after the stress procedure. These findings shed new light on the neurochemical changes that are involved in the compensatory response to adverse events in pubertal male rats and support other data that suggest a regulatory interplay between the RLN-3 pathway and the hypothalamus-pituitary-adrenal axis activity in the mechanisms of anxiety-like behavior.


Subject(s)
Anxiety , Brain , RNA, Messenger , Rats, Wistar , Receptors, G-Protein-Coupled , Stress, Psychological , Animals , Male , Rats , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Anxiety/metabolism , Anxiety/physiopathology , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Brain/metabolism , RNA, Messenger/metabolism , Behavior, Animal/physiology , Relaxin/metabolism , Relaxin/genetics , Receptors, Peptide/metabolism , Receptors, Peptide/genetics , Sexual Maturation/physiology , Nerve Tissue Proteins
4.
Brain Behav Immun ; 120: 315-326, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38852762

ABSTRACT

Social anxiety disorder is a common psychiatric condition that severely affects quality of life of individuals and is a significant societal burden. Although many risk factors for social anxiety exist, it is currently unknown how social fear sensitivity manifests biologically. Furthermore, since some individuals are resilient and others are susceptible to social fear, it is important to interrogate the mechanisms underpinning individual response to social fear situations. The microbiota-gut-brain axis has been associated with social behaviour, has recently been linked with social anxiety disorder, and may serve as a therapeutic target for modulation. Here, we assess the potential of this axis to be linked with social fear extinction processes in a murine model of social anxiety disorder. To this end, we correlated differential social fear responses with microbiota composition, central gene expression, and immune responses. Our data provide evidence that microbiota variability is strongly correlated with alterations in social fear behaviour. Moreover, we identified altered gene candidates by amygdalar transcriptomics that are linked with social fear sensitivity. These include genes associated with social behaviour (Armcx1, Fam69b, Kcnj9, Maoa, Serinc5, Slc6a17, Spata2, and Syngr1), inflammation and immunity (Cars, Ckmt1, Klf5, Maoa, Map3k12, Pex5, Serinc5, Sidt1, Spata2), and microbe-host interaction (Klf5, Map3k12, Serinc5, Sidt1). Together, these data provide further evidence for a role of the microbiota-gut-brain axis in social fear responses.


Subject(s)
Brain-Gut Axis , Extinction, Psychological , Fear , Gastrointestinal Microbiome , Mice, Inbred C57BL , Animals , Fear/physiology , Mice , Gastrointestinal Microbiome/physiology , Extinction, Psychological/physiology , Male , Brain-Gut Axis/physiology , Brain/metabolism , Social Behavior , Phobia, Social/metabolism , Phobia, Social/psychology , Amygdala/metabolism , Disease Models, Animal , Anxiety/metabolism
5.
Biochem Pharmacol ; 226: 116358, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38857830

ABSTRACT

With societal development and an ageing population, psychiatric disorders have become a common cause of severe and long-term disability and socioeconomic burdens worldwide. Semaphorin 3A (Sema-3A) is a secreted glycoprotein belonging to the semaphorin family. Sema-3A is well known as an axon guidance factor in the neuronal system and a potent immunoregulator at all stages of the immune response. It is reported to have various biological functions and is involved in many human diseases, including autoimmune diseases, angiocardiopathy, osteoporosis, and tumorigenesis. The signals of sema-3A involved in the pathogenesis of these conditions, are transduced through its cognate receptors and diverse downstream signalling pathways. An increasing number of studies show that sema-3A plays important roles in synaptic and dendritic development, which are closely associated with the pathophysiological mechanisms of psychiatric disorders, including schizophrenia, depression, and autism, suggesting the involvement of sema-3A in the pathogenesis of mental diseases. This indicates that mutations in sema-3A and alterations in its receptors and signalling may compromise neurodevelopment and predispose patients to these disorders. However, the role of sema-3A in psychiatric disorders, particularly in regulating neurodevelopment, remains elusive. In this review, we summarise the recent progress in understanding sema-3A in the pathogenesis of mental diseases and highlight sema-3A as a potential target for the prevention and treatment of these diseases.


Subject(s)
Schizophrenia , Semaphorin-3A , Humans , Semaphorin-3A/metabolism , Semaphorin-3A/genetics , Semaphorin-3A/physiology , Animals , Schizophrenia/metabolism , Schizophrenia/genetics , Depression/metabolism , Anxiety/metabolism , Mental Disorders/metabolism , Mental Disorders/genetics , Signal Transduction/physiology
6.
Nat Commun ; 15(1): 5439, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937485

ABSTRACT

Efficient control of feeding behavior requires the coordinated adjustment of complex motivational and affective neurocircuits. Neuropeptides from energy-sensing hypothalamic neurons are potent feeding modulators, but how these endogenous signals shape relevant circuits remains unclear. Here, we examine how the orexigenic neuropeptide Y (NPY) adapts GABAergic inputs to the bed nucleus of the stria terminalis (BNST). We find that fasting increases synaptic connectivity between agouti-related peptide (AgRP)-expressing 'hunger' and BNST neurons, a circuit that promotes feeding. In contrast, GABAergic input from the central amygdala (CeA), an extended amygdala circuit that decreases feeding, is reduced. Activating NPY-expressing AgRP neurons evokes these synaptic adaptations, which are absent in NPY-deficient mice. Moreover, fasting diminishes the ability of CeA projections in the BNST to suppress food intake, and NPY-deficient mice fail to decrease anxiety in order to promote feeding. Thus, AgRP neurons drive input-specific synaptic plasticity, enabling a selective shift in hunger and anxiety signaling during starvation through NPY.


Subject(s)
Agouti-Related Protein , Feeding Behavior , Neuronal Plasticity , Neuropeptide Y , Septal Nuclei , Starvation , Animals , Neuropeptide Y/metabolism , Neuropeptide Y/genetics , Neuronal Plasticity/physiology , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics , Feeding Behavior/physiology , Septal Nuclei/metabolism , Septal Nuclei/physiology , Mice , Starvation/metabolism , Male , Amygdala/metabolism , Amygdala/physiology , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/physiology , GABAergic Neurons/metabolism , Eating/physiology , Fasting/physiology , Anxiety/metabolism , Anxiety/physiopathology , Hunger/physiology
7.
Neuropharmacology ; 257: 110033, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38866066

ABSTRACT

The anteroventral bed nucleus of stria terminalis (avBNST) is a limbic forebrain region involved in the regulation of anxiety, and expresses GABAB receptors, which are located at both pre- and post-synaptic sites. However, it is unclear how blockade of these receptors affects anxiety-like behaviors, particularly in Parkinson's disease (PD)-related anxiety. In the present study, unilateral 6-hydroxydopamine (6-OHDA) lesions of the substantia nigra pars compacta in rats induced anxiety-like behaviors, and increased GABA release and decreased glutamate release in the avBNST, as well as decreased level of dopamine (DA) in the basolateral amygdala (BLA). Intra-avBNST injection of pre-synaptic GABAB receptor antagonist CGP36216 produced anxiolytic-like effects, while the injection of post-synaptic GABAB receptor antagonist CGP35348 induced anxiety-like responses in both sham and 6-OHDA rats. Intra-avBNST injection of CGP36216 inhibited the GABAergic neurons and increased GABA/glutamate ratio in the avBNST and increased levels of DA and serotonin (5-HT) in the BLA; conversely, CGP35348 produced opposite effects on the firing activity of avBNST GABAergic neurons and levels of the neurotransmitters in the avBNST and BLA. Moreover, the doses of the antagonists producing significant behavioral effects in 6-OHDA rats were lower than those in sham rats, and the duration of action of the antagonists on the firing rate of the neurons and release of the neurotransmitters was prolonged in 6-OHDA rats. Altogether, these findings suggest that pre- and post-synaptic GABAB receptors in the avBNST are implicated in PD-related anxiety-like behaviors, and degeneration of the nigrostriatal pathway enhances functions and/or upregulates expression of these receptors.


Subject(s)
Anti-Anxiety Agents , Anxiety , GABA-B Receptor Antagonists , Oxidopamine , Parkinsonian Disorders , Receptors, GABA-B , Septal Nuclei , Animals , Septal Nuclei/drug effects , Septal Nuclei/metabolism , Male , Anxiety/metabolism , GABA-B Receptor Antagonists/pharmacology , Anti-Anxiety Agents/pharmacology , Rats , Receptors, GABA-B/metabolism , Oxidopamine/toxicity , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/psychology , Dopamine/metabolism , Glutamic Acid/metabolism , gamma-Aminobutyric Acid/metabolism , Rats, Sprague-Dawley , Serotonin/metabolism , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/drug effects , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Organophosphorus Compounds
8.
Neuropharmacology ; 257: 110031, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38871116

ABSTRACT

Exposure to chronic and unpredictable stressors can precipitate mood-related disorders in humans, particularly in individuals with pre-existing mental health challenges. L-type calcium channels (LTCCs) have been implicated in numerous neuropsychiatric disorders, as LTCC encoding genes have been identified as candidate risk factors for neuropsychiatric illnesses. In these sets of experiments, we sought to examine the ability of LTCC blockade to alter depression, anxiety, and anhedonic-related behavioral responses to chronic unpredictable stress (CUS) exposure in female and male rats. Rats first underwent either 21 days of CUS or no exposure to chronic stressors, serving as home cage controls (HCC). Then rats were examined for anhedonia-related behavior, anxiety and depression-like behavioral responses as measured by the sucrose preference test (SPT), elevated plus maze (EPM), and forced swim test (FST). CUS exposed females and males showed anhedonic and anxiogenic-like behavioral responses on the SPT and EPM, respectively, when compared to HCCs. In female and male rats, systemic administration of the LTCC blocker isradipine (0.4 mg/kg and 1.2 mg/kg, I.P.) attenuated the CUS-induced decrease in sucrose preference and reversed the CUS-induced decrease in open arm time. In the FST, systemic isradipine decreased immobility time across all groups, consistent with an antidepressant-like response. However, there were no significant differences in forced swim test immobility time between HCC and CUS exposed animals. Taken together, these data point to a role of LTCCs in the regulation of mood disorder-related behavioral phenotype responses to chronic stress exposure.


Subject(s)
Anhedonia , Anxiety , Calcium Channel Blockers , Calcium Channels, L-Type , Depression , Stress, Psychological , Animals , Anhedonia/physiology , Anhedonia/drug effects , Male , Stress, Psychological/metabolism , Stress, Psychological/psychology , Female , Calcium Channels, L-Type/metabolism , Depression/metabolism , Anxiety/metabolism , Rats , Calcium Channel Blockers/pharmacology , Rats, Sprague-Dawley , Disease Models, Animal , Phenotype , Food Preferences/drug effects , Food Preferences/physiology
9.
FASEB J ; 38(11): e23648, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38822661

ABSTRACT

Previous studies on germ-free (GF) animals have described altered anxiety-like and social behaviors together with dysregulations in brain serotonin (5-HT) metabolism. Alterations in circulating 5-HT levels and gut 5-HT metabolism have also been reported in GF mice. In this study, we conducted an integrative analysis of various behaviors as well as markers of 5-HT metabolism in the brain and along the GI tract of GF male mice compared with conventional (CV) ones. We found a strong decrease in locomotor activity, accompanied by some signs of increased anxiety-like behavior in GF mice compared with CV mice. Brain gene expression analysis showed no differences in HTR1A and TPH2 genes. In the gut, we found decreased TPH1 expression in the colon of GF mice, while it was increased in the cecum. HTR1A expression was dramatically decreased in the colon, while HTR4 expression was increased both in the cecum and colon of GF mice compared with CV mice. Finally, SLC6A4 expression was increased in the ileum and colon of GF mice compared with CV mice. Our results add to the evidence that the microbiota is involved in regulation of behavior, although heterogeneity among studies suggests a strong impact of genetic and environmental factors on this microbiota-mediated regulation. While no impact of GF status on brain 5-HT was observed, substantial differences in gut 5-HT metabolism were noted, with tissue-dependent results indicating a varying role of microbiota along the GI tract.


Subject(s)
Behavior, Animal , Germ-Free Life , Serotonin , Animals , Serotonin/metabolism , Mice , Male , Gastrointestinal Microbiome/physiology , Brain/metabolism , Tryptophan Hydroxylase/metabolism , Tryptophan Hydroxylase/genetics , Anxiety/metabolism , Anxiety/microbiology , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Mice, Inbred C57BL , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/genetics , Colon/metabolism , Colon/microbiology
10.
PLoS One ; 19(6): e0304261, 2024.
Article in English | MEDLINE | ID: mdl-38870197

ABSTRACT

PURPOSE: Patients with Retinitis Pigmentosa (RP) commonly experience sleep-related issues and are susceptible to stress. Moreover, variatiaons in their vision are often linked to anxiety, stress and drowsiness, indicating that stress and sleep deprivation lead to a decline in vision, and vision improves when both are mitigated. The objective of this study was to investigate the utility of salivary biomarkers as biochemical indicators of anxiety and sleep deprivation in RP patients. METHODS: Seventy-eight RP patients and 34 healthy controls were included in this observational study. Anxiety and sleep-quality questionnaires, a complete ophthalmological exam for severity grading and, the collection of salivary samples from participants were assessed for participants. The activity of biomarkers was estimated by ELISA, and statistical analysis was performed to determine associations between the parameters. Associations between underlying psychological factors, grade of disease severity, and biomarkers activity were also examined. RESULTS: Fifty-two (67%) of patients had a severe RP, and 26 (33%) had a mild-moderate grade. Fifty-eight (58,9%) patients reported severe levels of anxiety and 18 (23.,1%) a high level. Forty-six (59%) patients obtained pathological values in sleep-quality questionaries and 43 (55.1%) in sleepiness. Patients with RP exhibited significant differences in testosterone, cortisol, sTNFαRII, sIgA and melatonin as compared to controls and patients with a mild-moderate and advanced stage of disease showed greater differences. In covariate analysis, patients with a severe anxiety level also showed greater differences in mean salivary cortisol, sTNFαRII and melatonin and male patients showed lower IgA levels than female. CONCLUSIONS: The present findings suggest that salivary biomarkers could be suitable non-invasive biochemical markers for the objective assessment of sleep deprivation and anxiety in RP patients. Further research is needed to characterize the effects of untreated negative psychological states and sleep deprivation on increased variability of vision and disease progression, if any.


Subject(s)
Biomarkers , Retinitis Pigmentosa , Saliva , Sleep Deprivation , Humans , Male , Female , Saliva/chemistry , Saliva/metabolism , Biomarkers/metabolism , Biomarkers/analysis , Retinitis Pigmentosa/metabolism , Adult , Middle Aged , Sleep Deprivation/metabolism , Stress, Psychological/metabolism , Anxiety/metabolism , Case-Control Studies , Hydrocortisone/analysis , Hydrocortisone/metabolism
11.
Behav Brain Res ; 470: 115094, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38844057

ABSTRACT

Estrogen receptor (ER) activation by 17-ß estradiol (E2) can attenuate neuronal injury and behavioral impairments following global cerebral ischemia (GCI) in rodents. This study sought to further examine the discrete roles of ERs through characterization of the effects of selective ER activation on post-ischemic pro-inflammatory microglial activation, hippocampal neuronal injury, and anxiety-like behaviors. Forty-six ovariectomized (OVX) adult female Wistar rats received daily s.c injections (100 µg/kg/day) of propylpyrazole triol (PPT; ERα agonist), diarylpropionitrile (DPN; ERß agonist), G-1 (G-protein coupled ER agonist; GPER), E2 (activating all receptors), or vehicle solution (VEH) for 21 days. After final injection, rats underwent GCI via 4-vessel occlusion (n=8 per group) or sham surgery (n=6, vehicle injections). The Open Field Test (OFT), Elevated Plus Maze (EPM), and Hole Board Test (HBT) assessed anxiety-like behaviors. Microglial activation (Iba1, CD68, CD86) in the basolateral amygdala (BLA), CA1 of the hippocampus, and paraventricular nucleus of the hypothalamus (PVN) was determined 8 days post-ischemia. Compared to sham rats, Iba1 activation and CA1 neuronal injury were increased in all ischemic groups except DPN-treated rats, with PPT-treated ischemic rats also showing increased PVN Iba1-ir expression. Behaviorally, VEH ischemic rats showed slightly elevated anxiety in the EPM compared to sham counterparts, with no significant effects of agonists. While no changes were observed in the OFT, emotion regulation via grooming in the HBT was increased in G-1 rats compared to E2 rats. Our findings support selective ER activation to regulate post-ischemic microglial activation and coping strategies in the HBT, despite minimal impact on hippocampal injury.


Subject(s)
Anxiety , Brain Ischemia , CA1 Region, Hippocampal , Microglia , Phenols , Pyrazoles , Rats, Wistar , Animals , Female , Microglia/metabolism , Microglia/drug effects , Rats , Anxiety/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/drug effects , Brain Ischemia/metabolism , Pyrazoles/pharmacology , Phenols/pharmacology , Ovariectomy , Neurons/metabolism , Neurons/drug effects , Propionates/pharmacology , Propionates/administration & dosage , Behavior, Animal/drug effects , Behavior, Animal/physiology , Estradiol/pharmacology , Disease Models, Animal , Receptors, Estrogen/metabolism , Nitriles/pharmacology
12.
Neurosci Biobehav Rev ; 163: 105748, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38857667

ABSTRACT

Corticotropin-releasing factor (CRF, corticoliberin) is a neuromodulatory peptide activating the hypothalamic-pituitary-adrenal (HPA) axis, widely distributed in the central nervous system (CNS) in mammals. In addition to its neuroendocrine effects, CRF is essential in regulating many functions under physiological and pathophysiological conditions through CRF1 and CRF2 receptors (CRF1R, CRF2R). This review aims to present selected examples of the diverse and sometimes opposite effects of CRF and its receptor ligands in various pathophysiological states, including stress/anxiety, depression, and processes associated with brain injury. It seems interesting to draw particular attention to the fact that CRF and its receptor ligands exert different effects depending on the brain structures or subregions, likely stemming from the varied distribution of CRFRs in these regions and interactions with other neurotransmitters. CRFR-mediated region-specific effects might also be related to brain site-specific ligand binding and the associated activated signaling pathways. Intriguingly, different types of CRF molecules can also influence the diverse actions of CRF in the CNS.


Subject(s)
Anxiety , Corticotropin-Releasing Hormone , Receptors, Corticotropin-Releasing Hormone , Receptors, Corticotropin-Releasing Hormone/metabolism , Humans , Animals , Corticotropin-Releasing Hormone/metabolism , Anxiety/metabolism , Anxiety/physiopathology , Brain Injuries/metabolism , Brain Injuries/physiopathology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Depression/metabolism , Depression/physiopathology , Brain/metabolism , Brain/physiopathology
13.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38866499

ABSTRACT

Previous studies have shown that ligands that bind to sigma-2 receptor/TMEM97 (s2R/TMEM97), a transmembrane protein, have anxiolytic/antidepressant-like properties and relieve neuropathic pain-like effects in rodents. Despite medical interest in s2R/TMEM97, little affective and pain behavioral characterization has been done using transgenic mice, which limits the development of s2R/TMEM97 as a viable therapeutic target. Using wild-type (WT) and global Tmem97 knock-out (KO) mice, we sought to identify the contribution of Tmem97 in modulating affective and pain-like behaviors using a battery of affective and pain assays, including open field, light/dark preference, elevated plus maze, forced swim test, tail suspension test, and the mechanical sensitivity tests. Our results demonstrate that female Tmem97 KO mice show less anxiety-like and depressive-like behaviors in light/dark preference and tail suspension tests but not in an open field, elevated plus maze, and forced swim tests at baseline. We next performed spared nerve injury in WT and Tmem97 KO mice to assess the role of Tmem97 in neuropathic pain-induced anxiety and depression. WT mice, but not Tmem97 KO mice, developed a prolonged neuropathic pain-induced depressive-like phenotype when tested 10 weeks after nerve injury in females. Our results show that Tmem97 plays a role in modulating anxiety-like and depressive-like behaviors in naive animals with a significant change in the presence of nerve injury in female mice. Overall, these data demonstrate that Tmem97 could be a target to alleviate affective comorbidities of pain disorders.


Subject(s)
Depression , Membrane Proteins , Mice, Inbred C57BL , Mice, Knockout , Neuralgia , Receptors, sigma , Animals , Receptors, sigma/metabolism , Female , Neuralgia/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Depression/metabolism , Depression/etiology , Behavior, Animal/physiology , Mice , Anxiety/metabolism , Disease Models, Animal , Male
14.
Adv Neurobiol ; 35: 157-182, 2024.
Article in English | MEDLINE | ID: mdl-38874723

ABSTRACT

Pain, fear, stress, and anxiety are separate yet interrelated phenomena. Each of these concepts has an extensive individual body of research, with some more recent work focusing on points of conceptual overlap. The role of the endogenous opioid system in each of these phenomena is only beginning to be examined and understood. Research examining the ways in which endogenous opioids (e.g., beta-endorphin; ßE) may mediate the relations among pain, fear, stress, and anxiety is even more nascent. This chapter explores the extant evidence for endogenous opioid activity as an underpinning mechanism of these related constructs, with an emphasis on research examining ßE.


Subject(s)
Anxiety , Fear , Pain , Stress, Psychological , Animals , Humans , Anxiety/metabolism , beta-Endorphin/metabolism , Fear/physiology , Opioid Peptides/metabolism , Pain/psychology , Pain/metabolism , Stress, Psychological/metabolism
15.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892090

ABSTRACT

Fetal hypoxia and maternal stress frequently culminate in neuropsychiatric afflictions in life. To replicate this condition, we employed a model of prenatal severe hypoxia (PSH) during days 14-16 of rat gestation. Subsequently, both control and PSH rats at 3 months old were subjected to episodes of inescapable stress to induce learned helplessness (LH). The results of the open field test revealed an inclination towards depressive-like behavior in PSH rats. Following LH episodes, control (but not PSH) rats displayed significant anxiety. LH induced an increase in glucocorticoid receptor (GR) levels in extrahypothalamic brain structures, with enhanced nuclear translocation in the hippocampus (HPC) observed both in control and PSH rats. However, only control rats showed an increase in GR nuclear translocation in the amygdala (AMG). The decreased GR levels in the HPC of PSH rats correlated with elevated levels of hypothalamic corticotropin-releasing hormone (CRH) compared with the controls. However, LH resulted in a reduction of the CRH levels in PSH rats, aligning them with those of control rats, without affecting the latter. This study presents evidence that PSH leads to depressive-like behavior in rats, associated with alterations in the glucocorticoid system. Notably, these impairments also contribute to increased resistance to severe stressors.


Subject(s)
Anxiety , Depression , Glucocorticoids , Prenatal Exposure Delayed Effects , Receptors, Glucocorticoid , Animals , Rats , Female , Anxiety/metabolism , Pregnancy , Glucocorticoids/metabolism , Depression/metabolism , Depression/etiology , Receptors, Glucocorticoid/metabolism , Prenatal Exposure Delayed Effects/metabolism , Stress, Psychological/metabolism , Male , Corticotropin-Releasing Hormone/metabolism , Hippocampus/metabolism , Hypoxia/metabolism , Phenotype , Behavior, Animal , Helplessness, Learned , Disease Models, Animal , Amygdala/metabolism , Fetal Hypoxia/metabolism , Fetal Hypoxia/complications
16.
Transl Psychiatry ; 14(1): 239, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38834575

ABSTRACT

Prior research has identified differential protein expression levels of linker histone H1x within the ventral hippocampus (vHipp) of stress-susceptible versus stress-resilient mice. These mice are behaviorally classified based on their divergent responses to chronic social stress. Here, we sought to determine whether elevated vHipp H1x protein levels directly contribute to these diverging behavioral adaptations to stress. First, we demonstrated that stress-susceptible mice uniquely express elevated vHipp H1x protein levels following chronic stress. Given that linker histones coordinate heterochromatin compaction, we hypothesize that elevated levels of H1x in the vHipp may impede pro-resilience transcriptional adaptations and prevent development of the resilient phenotype following social stress. To test this, 8-10-week-old male C57BL/6 J mice were randomly assigned to groups undergoing 10 days of chronic social defeat stress (CSDS) or single housing, respectively. Following CSDS, mice were classified as susceptible versus resilient based on their social interaction behaviors. We synthesized a viral overexpression (OE) vector for H1x and transduced all stressed and single housed mice with either H1x or control GFP within vHipp. Following viral delivery, we conducted social, anxiety-like, and memory-reliant behavior tests on distinct cohorts of mice. We found no behavioral adaptations following H1x OE compared to GFP controls in susceptible, resilient, or single housed mice. In sum, although we confirm elevated vHipp protein levels of H1x associate with susceptibility to social stress, we observe no significant behavioral consequence of H1x OE. Thus, we conclude elevated levels of H1x are associated with, but are not singularly sufficient to drive development of behavioral adaptations to stress.


Subject(s)
Behavior, Animal , Hippocampus , Histones , Mice, Inbred C57BL , Stress, Psychological , Animals , Male , Hippocampus/metabolism , Mice , Stress, Psychological/metabolism , Histones/metabolism , Behavior, Animal/physiology , Adaptation, Psychological/physiology , Resilience, Psychological , Social Defeat , Anxiety/metabolism
17.
Exp Brain Res ; 242(8): 1871-1879, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38864869

ABSTRACT

This study aimed to compare the effects of High-Intensity Interval Training (HIIT) performed in a single session(1xHIIT) versus three daily sessions (3xHIIT) on fitness level and behavior of aged rats. Eighteen-month-old Wistar rats were assigned to Untrained (UN), 1xHIIT, or 3xHIIT (n = 12/group). Both groups, 1xHIIT and 3xHIIT, performed 15 min of a treadmill running HIIT protocol during 8 weeks. 1xHIIT protocol consisted of a single daily session of 15 min, while the 3xHIIT performed three daily sessions of 5 min with a 4 h interval between the sessions. Morris Water Maze (MWM) task was used to evaluate spatial learning and memory. Splash test, Forced Swim test, and Elevated Plus Maze task (EPM) were used to evaluate anhedonic, depressive-like, and anxious behaviors, respectively. Rats were euthanized, and the hippocampus was harvested for western blot analyses (CaMKII and BDNF). Both HIIT protocols improved VO2max and spatial memory. Notably, only the 3xHIIT protocol attenuated anxious and depressive-like behaviors. Western blot analyses of the hippocampus revealed that both HIIT protocols increased BDNF levels. BDNF levels were higher in the 3xHIIT when compared with 1xHIIT group, and we observed increasement of the CamKII levels just in the 3x HIIT group. Therefore, this study provides evidence indicating that accumulated HIIT sessions is more effective than traditional daily HIIT sessions in improving fitness level, cognitive function, memory, inhibiting the development of mood disorders, and enhancing BDNF and CaMKII levels in the hippocampus of aged rats.


Subject(s)
Aging , Anxiety , Brain-Derived Neurotrophic Factor , Depression , High-Intensity Interval Training , Hippocampus , Rats, Wistar , Animals , Hippocampus/metabolism , Rats , Depression/metabolism , Depression/therapy , Depression/physiopathology , Aging/physiology , Aging/metabolism , Anxiety/metabolism , Anxiety/therapy , Anxiety/physiopathology , High-Intensity Interval Training/methods , Male , Brain-Derived Neurotrophic Factor/metabolism , Cognition/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Maze Learning/physiology , Physical Conditioning, Animal/physiology , Spatial Memory/physiology
18.
Neurosci Lett ; 836: 137862, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-38851448

ABSTRACT

The endocannabinoid system has been shown to be a powerful mediator of anxiety, learning and memory, as well as nociception behaviors. Exogenous cannabinoids like delta-9-tetrahydrocannabinol mimic the naturally occurring endogenous cannabinoids found in the mammalian central and peripheral nervous system. The hydrophobic properties of endocannabinoids mean that these psychoactive compounds require help with cellular transport. A family of lipid intracellular carriers called fatty acid-binding proteins (FABPs) can bind to endocannabinoids. Pharmacological inhibition or genetic deletion of FABP subtypes 5 and 7 elevates whole-brain anandamide (AEA) levels, a type of endocannabinoid. This study examined locomotor behavior, anxiety-like behavior, and social behavior in FABP5-/- and FABP7-/- mice. Furthermore, we measured N-methyl-D-aspartate (NMDA) receptor levels in the brain to help identify potential underlying mechanisms related to the behavioral findings. Results showed that both male and female FABP5-/- mice exhibited significantly lower activity when compared with both FABP5/7+/+ (control) and FABP7-/-. For social behavior, male, but not female, FABP5-/- mice spent more time interacting with novel mice compared with controls (FABP5/7+/+) and FABP7-/- mice. No significant difference was found for anxiety-like behavior. Results from the NMDA autoradiography revealed [3H] MK-801 binding to be significantly increased within sub-regions of the striatum in FABP7-/- compared with control. In summary, these results show that FABP5 deficiency plays a significant role in locomotion activity, exploratory behavior, as well as social interaction. Furthermore, FABP7 deficiency is shown to play an important role in NMDA receptor expression, while FABP5 does not.


Subject(s)
Anxiety , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Locomotion , Mice, Knockout , Receptors, N-Methyl-D-Aspartate , Social Behavior , Animals , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/genetics , Anxiety/metabolism , Male , Female , Fatty Acid-Binding Protein 7/metabolism , Locomotion/physiology , Mice , Receptors, N-Methyl-D-Aspartate/metabolism , Mice, Inbred C57BL , Signal Transduction , Brain/metabolism , N-Methylaspartate/metabolism , Neoplasm Proteins
19.
Front Endocrinol (Lausanne) ; 15: 1380779, 2024.
Article in English | MEDLINE | ID: mdl-38919481

ABSTRACT

Objective: Aromatherapy is a holistic healing method to promote health and well-being by using natural plant extracts. However, its precise mechanism of action and influence on the endocrine system remains unclear. Since recent studies reported that a neuropeptide, oxytocin, can attenuate anxiety, we hypothesized that if oxytocin secretion is promoted through aromatherapy, it may improve mood and anxiety. The present study is aimed to investigate the relationship between oxytocin and the effects of aromatherapy with lavender oil on anxiety level, by measuring salivary oxytocin levels in healthy men and women. Methods: We conducted a randomized open crossover trial in 15 men and 10 women. Each participant received a placebo intervention (control group) and aromatherapy with lavender oil (aromatherapy group). For the aromatherapy group, each participant spent a 30-min session in a room with diffused lavender essential oil, followed by a 10-min hand massage using a carrier oil containing lavender oil. Anxiety was assessed using the State-Trait Anxiety Inventory (STAI) before the intervention, 30-min after the start of intervention, and after hand massage, in both groups. Saliva samples were collected at the same time points of the STAI. Results: In women, either aromatherapy or hand massage was associated with a reduction in anxiety levels, independently. Moreover, salivary oxytocin levels were increased after aromatherapy. On the other hand, in men, anxiety levels were decreased after aromatherapy, as well as after hand massage, regardless of the use of lavender oil. However, there were no significant differences in changes of salivary oxytocin levels between the control and aromatherapy groups during the intervention period. Interestingly, there was a positive correlation between anxiety levels and salivary oxytocin levels before the intervention, but a negative correlation was observed after hand massage with lavender oil. Conclusion: The results of the present study indicate that in women, aromatherapy with lavender oil attenuated anxiety with increase in oxytocin level in women, whereas in men, there was no clear relationship of aromatherapy with anxiety or oxytocin levels but, there was a change in correlation between anxiety and oxytocin. The results of the present study suggest that the effect of aromatherapy can vary depending on sex.


Subject(s)
Anxiety , Aromatherapy , Cross-Over Studies , Lavandula , Oils, Volatile , Oxytocin , Plant Oils , Saliva , Humans , Oxytocin/metabolism , Aromatherapy/methods , Female , Male , Saliva/chemistry , Saliva/metabolism , Anxiety/therapy , Anxiety/metabolism , Adult , Oils, Volatile/therapeutic use , Lavandula/chemistry , Young Adult , Sex Characteristics
20.
Drug Des Devel Ther ; 18: 2227-2248, 2024.
Article in English | MEDLINE | ID: mdl-38882046

ABSTRACT

Purpose: The Baihe Dihuang decoction (BDD) is a representative traditional Chinese medicinal formula that has been used to treat anxiety disorders for thousands of years. This study aimed to reveal mechanisms of anxiolytic effects of BDD with multidimensional omics. Methods: First, 28-day chronic restraint stress (CRS) was used to create a rat model of anxiety, and the open field test and elevated plus maze were used to assess anxiety-like behavior. Enzyme-linked immunosorbent assay (ELISA), hematoxylin-eosin staining, and immunofluorescence staining were used to evaluate inflammatory response. Besides, 16S rRNA gene sequencing assessed fecal microbiota composition and differential microbiota. Non-targeted metabolomics analysis of feces was performed to determine fecal biomarkers, and targeted metabolomics was used to observe the levels of hippocampus neurotransmitters. Finally, Pearson correlation analysis was used to examine relationships among gut microbiota, fecal metabolites, and neurotransmitters. Results: BDD significantly improved anxiety-like behaviors in CRS-induced rats and effectively ameliorated hippocampal neuronal damage and abnormal activation of hippocampal microglia. It also had a profound effect on the diversity of microbiota, as evidenced by significant changes in the abundance of 10 potential microbial biomarkers at the genus level. Additionally, BDD led to significant alterations in 18 fecal metabolites and 12 hippocampal neurotransmitters, with the majority of the metabolites implicated in amino acid metabolism pathways such as D-glutamine and D-glutamate, alanine, arginine and proline, and tryptophan metabolism. Furthermore, Pearson analysis showed a strong link among gut microbiota, metabolites, and neurotransmitters during anxiety and BDD treatment. Conclusion: BDD can effectively improve anxiety-like behaviors by regulating the gut-brain axis, including gut microbiota and metabolite modification, suppression of hippocampal neuronal inflammation, and regulation of neurotransmitters.


Subject(s)
Anti-Anxiety Agents , Disease Models, Animal , Drugs, Chinese Herbal , Gastrointestinal Microbiome , Metabolomics , Rats, Sprague-Dawley , Animals , Rats , Anti-Anxiety Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Male , Gastrointestinal Microbiome/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Anxiety/drug therapy , Anxiety/metabolism , Restraint, Physical , Hippocampus/drug effects , Hippocampus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL