Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.995
Filter
1.
Phytomedicine ; 134: 155999, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39241390

ABSTRACT

BACKGROUND: Depression and anxiety disorders are prevalent psychiatric conditions, and currently utilized chemical drugs typically come with significant adverse effects. China boasts a wealth of medicinal and food herbs known for their safe and effective properties. PURPOSE: This study aimed to develop novel formulations with improved antidepressant and anxiolytic effects derived from medicinal and food herbs. STUDY DESIGN: Screening combinations with antidepressant and anxiolytic effects using techniques such as network pharmacology and validating their effects in vitro and in vivo experiments. METHODS: Utilizing network pharmacology and molecular docking, we identified the top ten medicinal herbs with anxiolytic and antidepressant potential. Herbs with cytoprotective effects and non-toxic characteristics were further screened to formulate the herbal blends. Subsequently, we established a PC12 cell injury model and a chronic unpredictable mild stress (CUMS) model in mice to assess the effects of our formulations. RESULTS: Ten medicinal herbs were initially screened, and six of them were deemed suitable for formulating the blend, namely Gancao, Dazao, Gouqizi, Sangye, Huangqi, and Jinyinhua (GDGSHJ). The GDGSHJ formulation reduced Lactate Dehydrogenase (LDH) leakage, decreased apoptosis, and demonstrated a favorable antidepressant and antianxiety effect in the CUMS mouse model. Besides, GDGSHJ led to the upregulation of serum 5-Hydroxytryptamine (5-HT) content and brain tissue 5-HT, Gamma-aminobutyric acid (GABA), and Dopamine (DA) levels. It also downregulated the expression of SLC6A4 and SLC6A3 genes in the mouse hippocampus while upregulating HTR1A, DRD1, DRD2, and GABRA1 genes. CONCLUSION: Our formulation exhibited robust antidepressant and antianxiety effects without inducing substantial toxicity. This efficacy appears to be mediated by the expression of relevant genes within the hippocampus of mice. The formulation achieved this effect by balancing 5-HT levels in the serum and DA, GABA, and 5-HT levels within brain tissue.


Subject(s)
Anti-Anxiety Agents , Antidepressive Agents , Molecular Docking Simulation , Network Pharmacology , Animals , Antidepressive Agents/pharmacology , Anti-Anxiety Agents/pharmacology , Mice , Male , PC12 Cells , Rats , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Depression/drug therapy , Plants, Medicinal/chemistry , Hippocampus/drug effects , Hippocampus/metabolism , Disease Models, Animal , Stress, Psychological/drug therapy , Anxiety/drug therapy , Serotonin/metabolism
2.
Chem Biol Drug Des ; 104(3): e14626, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39307842

ABSTRACT

Depression is a prevalent mental disorder, but the side effects of antidepressants also make depressed patients resistant. Effective and safe antidepressants should be developed from traditional herbs, with the aim of reducing the side effects of antidepressants and improving the efficacy of drugs. In this study, the new macamide compound-4 (NMC-4) was synthesized for the first time, addressing the problem of difficult extraction, isolation, and low content of natural macamide. NMC-4 was characterized using mass spectrometry, nuclear magnetic resonance, and infrared spectroscopy. The protective effect of NMC-4 against cell injury was demonstrated to be stronger than that of natural macamide (N-benzylhexadecanamide, XA) using a PC12 cell injury model. The study explored the effects of NMC-4 on chronic unpredictable mild stress (CUMS)-induced depressive symptoms. NMC-4 significantly improved depressive-like behaviors. NMC-4 ameliorated CUMS-induced depressive-like behaviors by mitigating neuroinflammation and modulating the NF-κB/Nrf2 and BDNF/PI3K/Akt pathways.


Subject(s)
Antidepressive Agents , Depression , Animals , Depression/drug therapy , Rats , PC12 Cells , Male , Antidepressive Agents/pharmacology , Antidepressive Agents/chemistry , Antidepressive Agents/therapeutic use , Stress, Psychological/drug therapy , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Signal Transduction/drug effects , Disease Models, Animal , NF-kappa B/metabolism , Mice , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , NF-E2-Related Factor 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
3.
Int J Med Mushrooms ; 26(11): 51-64, 2024.
Article in English | MEDLINE | ID: mdl-39241163

ABSTRACT

Psychological disparities impact physical activity and fitness in sedentary female college students by affecting cardiovascular efficiency. Ganoderma lucidum, vitality-enhancing herb alleviates health and rejuvenates the mind-body to improve endurance fitness. A double-blinded, randomized, placebo-controlled parallel design study was conducted to determine whether supplementation of G. lucidum in daily dosages of 500 mg (GL500mg group) and 1000 mg (GL1000mg group) improves psychophysiological health capabilities during the different phases of the experimental trial. Analysis for pre-experimental trial (day 0), experimental trial (day 15), and post-experimental trial (after day 30) on anthropometric, psychological, physiological, and physical fitness parameters were executed. Seventy-eight participants (n = 78, age 20.64 ± 3.21 years) were assigned randomly and equally divided (n = 26) to one of the three treatment groups for intragroup and intergroup comparisons. Significant differences in the post-experimental GL1000mg group for heart rate (HR), maximal oxygen consumption (VO2max), physical work capacity (PWC170), and right-hand grip strength (P < 0.05) compared with the placebo group were observed. GL1000mg-supplemented group also significantly improved (P < 0.05) HR, VO2max and PWC170 (P < 0.001) after pre- to post-trials. Experimental trial between placebo and GL1000mg group and post-experimental trial between the GL500mg and GL1000mg group showed significant changes in VO2max(P < 0.001) and PWC170 (P < 0.05). Anxiety, depression, vitality and positive well-being scores significantly improved, leading to improved psychological well-being after GL1000mg supplementation. GL1000mg supplementation for 30 days might act as a longevity-promoting tonic for endurance and strength performance by ameliorating stress to improve the overall psychophysiological health, vitality and quality of life for better cardiovascular efficacy.


Subject(s)
Dietary Supplements , Physical Fitness , Reishi , Stress, Psychological , Students , Humans , Female , Reishi/chemistry , Young Adult , Students/psychology , India , Double-Blind Method , Dietary Supplements/analysis , Stress, Psychological/drug therapy , Adult , Adolescent , Heart Rate/drug effects , Oxygen Consumption/drug effects , Hand Strength , Universities
4.
Behav Pharmacol ; 35(7): 408-417, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39230572

ABSTRACT

Omeprazole, a drug of choice for the management of gastric hyperacidity, influences serotonergic neurotransmission in brain regions and its long-term use is known to cause stress-related behavioral deficits including anxiety. Aim of the current study was to explore the effects of omeprazole treatment on immobilization-induced anxiety in rats, specifically on the role of serotonin (5-HT). In view of the role of serotonin-1A (5-HT1A) autoreceptor in the availability of 5-HT in brain regions, mRNA expression of this autoreceptor was performed in raphe nuclei. Similarly, because of the role of hippocampal 5-HT neurotransmission in anxiety-like disorders, expression of the 5-HT1A heteroreceptors was determined in this region. We found that the treatment with omeprazole reduces anxiety-like behavior in rats, increases the expression of 5-HT1A autoreceptor in the raphe and decreases the hippocampal expression of 5-HT1A heteroreceptor. This suggests a role of 5-HT1A receptor types in omeprazole-induced behavioral changes. It also indicates a potential role of omeprazole in the management of serotonergic disorders.


Subject(s)
Anxiety , Disease Models, Animal , Hippocampus , Omeprazole , Receptor, Serotonin, 5-HT1A , Stress, Psychological , Animals , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/drug effects , Omeprazole/pharmacology , Male , Rats , Anxiety/drug therapy , Anxiety/metabolism , Stress, Psychological/metabolism , Stress, Psychological/drug therapy , Hippocampus/metabolism , Hippocampus/drug effects , Rats, Wistar , Brain/metabolism , Brain/drug effects , Serotonin/metabolism , Raphe Nuclei/metabolism , Raphe Nuclei/drug effects , RNA, Messenger/metabolism , Restraint, Physical , Immobilization
5.
Nutrients ; 16(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39275174

ABSTRACT

The medicinal properties of resveratrol have garnered increasing attention from researchers. Extensive data have been accumulated on its use in treating cardiovascular diseases, immune system disorders, cancer, neurological diseases, and behavioral disorders. The protective mechanisms of resveratrol, particularly in anxiety-related stress disorders, have been well documented. However, less attention has been given to the side effects of resveratrol. This review explores not only the mechanisms underlying the anxiolytic effects of resveratrol but also the mechanisms that may lead to increased anxiety following resveratrol treatment. Understanding these mechanisms is crucial for enhancing the efficacy of resveratrol in managing anxiety disorders associated with stress and PTSD.


Subject(s)
Anti-Anxiety Agents , Anxiety Disorders , Anxiety , Resveratrol , Resveratrol/pharmacology , Humans , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Animals , Anxiety/drug therapy , Anxiety Disorders/drug therapy , Stress, Psychological/drug therapy , Stress Disorders, Post-Traumatic/drug therapy
6.
Langmuir ; 40(37): 19739-19750, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39219094

ABSTRACT

Depression is a debilitating mental illness that severely threatens millions of individuals and public health. Because of the multifactorial etiologies, there is currently no cure for depression; thus, it is urgently imperative to find alternative antidepressants and strategies. Growing evidence underscores the prominent role of oxidative stress as key pathological hallmarks of depression, making oxidative stress a potential therapeutic target. In this study, we report a N-doped carbon dot nanozyme (CDzyme) with excellent antioxidant capacity for treating depression by remodeling redox homeostasis and gut microbiota. The CDzymes prepared via microwave-assisted fast polymerization of histidine and glucose exhibit superior biocompatibility. Benefiting from the unique structure, CDzymes can provide abundant electrons, hydrogen atoms, and protons for reducing reactions, as well as catalytic sites to mimic redox enzymes. These mechanisms collaborating endow CDzymes with broad-spectrum antioxidant capacity to scavenge reactive oxygen and nitrogen species (•OH, O2-•, H2O2, ONOO-), and oxygen/nitrogen centered free radicals. A depression animal model was established by chronic unpredictable mild stress (CUMS) to evaluate the therapeutic efficacy of CDzymes from the behavioral, physiological, and biochemical index and intestinal flora assessments. CDzymes can remarkably improve depression-like behaviors and key neurotransmitters produced in hippocampus tissues and restore the gut microbiota compositions and the amino acid metabolic functions, proving the potential in treating depression through the intestinal-brain axis system. This study will facilitate the development of intestinal flora dysbiosis nanomedicines and treatment strategies for depression and other oxidative stress related multifactorial diseases.


Subject(s)
Antioxidants , Carbon , Depression , Gastrointestinal Microbiome , Animals , Gastrointestinal Microbiome/drug effects , Carbon/chemistry , Carbon/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Depression/drug therapy , Mice , Oxidative Stress/drug effects , Stress, Psychological/drug therapy , Male , Quantum Dots/chemistry , Antidepressive Agents/pharmacology , Antidepressive Agents/chemistry
7.
BMC Complement Med Ther ; 24(1): 339, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304871

ABSTRACT

BACKGROUND: Gegen Qinlian Decoction (GQD) is a classical traditional Chinese medicine (TCM) formula primarily utilized for treating gut disorders. GQD showed therapeutic effects on several diseases in clinical and animal studies by targeting gut microbes. Our recent studies also found that GQD efficiently alleviated anxiety in methamphetamine-withdrawn mice via regulating gut microbiome and metabolism. Given that various studies have indicated the link between the gut microbiome and the development of depression, here we endeavor to explore whether GQD can manage depression disorders by targeting the gut microbiome. METHODS AND MATERIALS: The depression-like model was induced in rats through chronic unpredictable mild stress (CUMS) and the depression levels were determined using the sucrose preference test (SPT). To address the depression-like behavior in rats, oral administration of GQD was employed. The colon microbiome and metabolite patterns were determined by 16s rRNA sequencing and untargeted metabolomics, respectively. RESULTS: We found 6 weeks of CUMS can induce depression-like behavior in rats and 4 weeks of GQD treatment can significantly alleviate the depression-like behavior. GQD treatment can also ameliorate the histological lesions in the colon of CUMS rats. Then, CUMS increased the abundance of gut microbes, while GQD treatment can restore it to a lower level. We further discovered that the abundances of 19 bacteria at the genus level were changed with CUMS treatment, among which the abundances of Ruminococcus, Lachnoclostridium, Pygmaiobacter, Bacteroides, Pseudomonas, and Pseudomonas Family_XIII_AD3011_group were stored by GQD treatment. Besides, we identified the levels of 36 colon metabolites were changed with CUMS treatment, among which the levels of Fasciculic acid B, Spermine, Fludrocortisone acetate, alpha-Ketoglutaric acid, 2-Oxoglutaric acid, N'-(benzoyloxy)-2-(2,2-dichlorocyclopropyl) ethanimidamide, N6-Succinyl Adenosine Oleanolic acid, KQH, Ergosta-5,7,9(11),22-Tetraen-3-beta-Ol, Gentisic acid, 4-Hydroxyretinoic Acid, FAHFA (3:0/16:0), Leucine-enkephalin and N-lactoyl-phenylalanine can be restored by GQD treatment. CONCLUSION: Our findings provide evidence supporting the therapeutic efficacy of GQD in alleviating depression-like behavior in CUMS rats, potentially being targeted on colon bacteria (especially the abundance of Ruminococcus and Bacteroides) and metabolites (especially the level of Oleanolic acid).


Subject(s)
Depression , Disease Models, Animal , Drugs, Chinese Herbal , Gastrointestinal Microbiome , Rats, Sprague-Dawley , Animals , Gastrointestinal Microbiome/drug effects , Rats , Drugs, Chinese Herbal/pharmacology , Male , Depression/drug therapy , Stress, Psychological/drug therapy , Behavior, Animal/drug effects
8.
J Ethnopharmacol ; 335: 118683, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39121928

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Depression impairs not only central nervous system, but also peripheral systems of the host. Gut microbiota have been proved to be involved in the pathogenesis of depression. Xiaoyaosan (XYS) has a history of over a thousand years in China for treating depression, dramatically alleviating anxiety, cognitive disorders, and especially gastrointestinal dysfunctions. Yet, it still just scratches the surface of the anti-depression mechanisms of XYS. AIM OF THE STUDY: This study aims to elucidate the mechanism of actions of XYS from the perspective of "microbiota-gut-brain" axis. MATERIALS AND METHODS: We firstly evaluated the effects of XYS on the macroscopic behaviors of depressed rats that induced by chronic unpredictable mild stress (CUMS). Secondly, the effects of XYS on intestinal homeostasis of depressed rats were revealed by using dysbacteriosis model. Subsequently, the underlying mechanisms were demonstrated by 16S rRNA gene sequencing technology and molecular biology methods. Finally, correlation analysis and visualization of the anti-depression effects of XYS were performed from the "microbiota - gut - brain" perspective. RESULTS: Our data indicated that XYS ameliorated the depression-like symptoms of CUMS rats, partly depending on the presence of gut microbiota. Furthermore, we illustrated that XYS reversed CUMS-induced gut dysbiosis of depressed rats in terms of decreasing the Bacteroidetes/Firmicutes ratio and the abundances of Bacteroides, and Corynebacterium, while increasing the abundances of Lactobacillus and Adlercreutzia. The significant enrichment of Bacteroides and the level of lipopolysaccharides (LPS) suggested that depression damaged the immune responses and gut barrier. Mechanistically, XYS significantly down-regulated the expression levels of factors that involved in TLR4/NLRP3 signaling pathway in the colon and brain tissues of depressed rats. In addition, XYS significantly increased the levels of claudin 1 and ZO-1, showing that XYS positively maintained the integrity of gut and blood-brain barriers (BBB). CONCLUSION: Our study offers insights into the anti-depression effects of XYS through a lens of "microbiota-TLR4/NLRP3 signaling pathway-barriers", providing a foundation for enhancing clinical efficiency and enriching drug selection, and contributing to our understanding of the mechanisms of traditional Chinese medicines (TCMs) in treating depression.


Subject(s)
Antidepressive Agents , Brain-Gut Axis , Depression , Drugs, Chinese Herbal , Gastrointestinal Microbiome , Lipopolysaccharides , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptor 4 , Animals , Gastrointestinal Microbiome/drug effects , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Male , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Depression/drug therapy , Depression/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats , Brain-Gut Axis/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Stress, Psychological/drug therapy , Stress, Psychological/psychology , Stress, Psychological/metabolism , Dysbiosis/drug therapy , Brain/drug effects , Brain/metabolism , Disease Models, Animal
9.
J Ethnopharmacol ; 335: 118663, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39128797

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Depression is a prevalent stress disorder, yet the underlying physiological mechanisms linking stress to appetite and weight loss remain elusive. While most antidepressants are associated with excessive weight and appetite gain, sertraline (SER) exhibits a lower risk of these side effects. Metacinnabar (ß-HgS), the primary component of Tibetan medicine Zuotai, has been shown to enhance mice's resilience against external stress without causing excessive increases in weight or appetite. However, the precise physiological pathway through which ß-HgS restores appetite and weight in stressed mice remains unclear. AIM OF THE STUDY: The objective of this study is to assess the efficacy of ß-HgS in ameliorating weight loss and appetite suppression induced by pressure stimulation in mice, as well as elucidate its potential mechanisms of action. METHODS: The present study employed chronic restraint stress (CRS) and chronic unpredictable mild stress (CUMS) as experimental models to simulate environmental stress encountered in daily life. Subsequently, a series of experiments were conducted, including behavior tests, HE staining of rectal and hippocampal pathological sections, detection of depression-related biological indicators, analysis of intestinal flora diversity, as well as metabolomics analysis of hippocampal and intestinal contents. RESULT: Dysregulation of glycerophospholipid metabolism may represent the principal pathway underlying reduced appetite, body weight, neurotransmitter and appetite hormone levels, heightened inflammatory response, hippocampal and rectal tissue damage, as well as altered composition of intestinal microbiota in stressed mice. Following intervention with SER and ß-HgS in stressed mice, the deleterious effects induced by stress can be ameliorated, in which the medium-dose ß-HgS exhibited superior performance. CONCLUSION: The aforementioned research findings suggest that the stress-induced decrease in appetite and body weight in mice may be associated with dysregulation in glycerophospholipid metabolism connecting the gut-brain axis. ß-HgS exhibits potential in ameliorating depressive-like symptoms in mice subjected to stress, while concurrently restoring their body weight and appetite without inducing excessive augmentation. Its therapeutic effect may also be attributed to its ability to modulate glycerophospholipid metabolism status and exert influence on the gut-brain axis.


Subject(s)
Appetite , Gastrointestinal Microbiome , Stress, Psychological , Animals , Male , Stress, Psychological/drug therapy , Mice , Appetite/drug effects , Gastrointestinal Microbiome/drug effects , Body Weight/drug effects , Depression/drug therapy , Antidepressive Agents/pharmacology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Behavior, Animal/drug effects
10.
Int Immunopharmacol ; 141: 113011, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39213872

ABSTRACT

Depression is a serious mental disorder that threatens patients' physical and mental health worldwide. The activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome is essential for microglia-mediated neuroinflammation and neuronal damage in depression. Numerous pathophysiological factors, such as mitochondrial dysfunction and impaired mitophagy, have an essential role in activating the NLRP3 inflammasome. AdipoRon is a potent adiponectin receptor agonist; however, its antidepressant effects have not been thoroughly investigated. In this study, we found that AdipoRon ameliorated depression-like behavior and neuronal damage induced by chronic unpredictable mild stress (CUMS). Further research demonstrated that AdipoRon inhibited the activation of the NLRP3 inflammasome and protected hippocampal neurons from microglial cytotoxicity by promoting mitophagy, increasing the clearance of damaged mitochondria, and reducing mtROS accumulation. Importantly, inhibition of mitophagy attenuated the antidepressant and neuroprotective effects of AdipoRon. Overall, these findings indicate that AdipoRon alleviates depression by inhibiting NLRP3 inflammasome activation in microglia via improving mitophagy.


Subject(s)
Antidepressive Agents , Depression , Inflammasomes , Mice, Inbred C57BL , Microglia , Mitophagy , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Mitophagy/drug effects , Microglia/drug effects , Microglia/metabolism , Inflammasomes/metabolism , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depression/drug therapy , Depression/metabolism , Male , Mice , Neurons/drug effects , Neurons/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Disease Models, Animal , Humans , Piperidines/pharmacology , Piperidines/therapeutic use , Stress, Psychological/drug therapy , Stress, Psychological/immunology , Cells, Cultured
11.
Int Immunopharmacol ; 141: 112985, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39213873

ABSTRACT

BACKGROUND: Depression is a common mental illness with more than 280 million sufferers worldwide. Inflammation, particularly the c-Jun amino-terminal kinase (JNK) pathway, contributes to depression development and neuronal apoptosis. Gardenia is a herb with therapeutic effects on depression that has been shown to inhibit neuronal apoptosis. However, one of the components in gardenia, Genipin 1-O-ß-D-gentiobioside(GG), has been less studied for its mechanism on depression. Thus, in the current study, we investigate how Genipin 1-O-ß-D-gentiobioside improves depression and elucidate its possible mechanism of action. METHODS: In this investigation, we utilize a chronic unpredictable mild stress (CUMS) mouse model and corticosterone-induced primary cortical neurons to examine the role of GG in ameliorating depressive symptoms and neuronal apoptosis. TUNEL staining and flow cytometry assessed the effects of GG on neuronal apoptosis. Western Blot analyses and immunofluorescence assays apoptosis-related proteins in the prefrontal cortex and primary neurons. The site of action of GG in regulating homeodomain interacting protein kinase 2 (HIPK2) SUMOylation was further explored in primary neurons. We constructed siRNA-SUMO1 vectors to transfect primary neuronal cells with intracellular SUMO1 knockdown. Proximity ligation assay (PLA) experiments were performed on primary neurons according to the instructions of the assay kit to observe the physical relationship between HIPK2 and SUMO1. We predicted the HIPK2 SUMOylation modification site by an online database and constructed vectors to target and site-directed mutagenesis, then to transfected primary neuronal cells. RESULTS: The results showed that GG effectively alleviated depressive-like behaviours, down-regulated apoptosis-related proteins (p-JNK, Bax, Cleaved-Caspase-3), and inhibited neuronal apoptosis in CUMS-induced depressed mice and corticosterone-induced primary cortical neurons. We reveal a complex mechanism underlying the link between GG, SUMOylation of HIPK2, and complex pathways of neuronal apoptosis regulation. K326 and K1189 are the key SUMOylation sites regulated by GG in intricate interactions of apoptosis-related proteins. CONCLUSION: Our study demonstrated that GG exerts antidepressant-like actions through neuroprotective effects by inhibiting the apoptosis of prefrontal cortex neurons, revealing the mechanism of GG inhibition of JNK phosphorylation by enhancing HIPK2 SUMOylation.


Subject(s)
Apoptosis , Depression , Mice, Inbred C57BL , Neurons , Prefrontal Cortex , Protein Serine-Threonine Kinases , Sumoylation , Animals , Neurons/drug effects , Apoptosis/drug effects , Sumoylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Male , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Mice , Depression/drug therapy , Depression/metabolism , Cells, Cultured , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Disease Models, Animal , Carrier Proteins/metabolism , Carrier Proteins/genetics , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Iridoids/pharmacology , Iridoids/therapeutic use , Iridoid Glucosides/pharmacology , Iridoid Glucosides/therapeutic use , Corticosterone
12.
Int J Mol Sci ; 25(15)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39125857

ABSTRACT

Neoponcirin causes anxiolytic-like effects in mice when administered intraperitoneally but not orally. Neoponcirin is non-water-soluble and insoluble in solvents, and in medium acid, it isomerizes, reducing its bioavailability. To improve the pharmacological properties of neoponcirin, we formed a neoponcirin complex with beta-cyclodextrin (NEO/ßCD), which was characterized by FT-IR, UV-Vis, and NMR, and their solubility profile. We evaluated the antidepressant-like effects of NEO/ßCD acutely administered to mice orally in the behavioral paradigms, the tail suspension (TST) and the forced swimming (FST) tests. We also analyzed the benefits of repeated oral doses of NEO/ßCD on depression- and anxiety-like behaviors induced in mice by chronic unpredictable mild stress (CUMS), using the FST, hole board, and open field tests. We determined the stressed mice's expression of stress-related inflammatory cytokines (IL-1ß, IL-6, and TNFα) and corticosterone. Results showed that a single or chronic oral administration of NEO/ßCD caused a robust antidepressant-like effect without affecting the ambulatory activity. In mice under CUMS, NEO/ßCD also produced anxiolytic-like effects and avoided increased corticosterone and IL-1ß levels. The effects of the NEO/ßCD complex were robust in both the acute and the stress chronic models, improving brain neurochemistry and recovering immune responses previously affected by prolonged stress.


Subject(s)
Antidepressive Agents , Depression , Stress, Psychological , beta-Cyclodextrins , Animals , beta-Cyclodextrins/pharmacology , beta-Cyclodextrins/chemistry , Mice , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Male , Stress, Psychological/drug therapy , Depression/drug therapy , Behavior, Animal/drug effects , Cytokines/metabolism , Disease Models, Animal , Anxiety/drug therapy , Anti-Anxiety Agents/pharmacology , Swimming , Administration, Oral
13.
Behav Pharmacol ; 35(6): 314-326, 2024 09 01.
Article in English | MEDLINE | ID: mdl-39094014

ABSTRACT

Depression is a common mood disorder and many patients do not respond to conventional pharmacotherapy or experience a variety of adverse effects. This work proposed that riparin I (RIP I) and riparin II (RIP II) present neuroprotective effects through modulation of astrocytes and microglia, resulting in the reversal of depressive-like behaviors. To verify our hypothesis and clarify the pathways underlying the effect of RIP I and RIP II on neuroinflammation, we used the chronic unpredictable mild stress (CUMS) depression model in mice. Male Swiss mice were exposed to stressors for 28 days. From 15 th to the 22 nd day, the animals received RIP I or RIP II (50 mg/kg) or fluoxetine (FLU, 10 mg/kg) or vehicle, by gavage. On the 29 th day, behavioral tests were performed. Expressions of microglia (ionized calcium-binding adaptor molecule-1 - Iba-1) and astrocyte (glial fibrillary acidic protein - GFAP) markers and levels of cytokines tumor necrosis factor alfa (TNF-α) and interleukin 1 beta (IL-1ß) were measured in the hippocampus. CUMS induced depressive-like behaviors and cognitive impairment, high TNF-α and IL-1ß levels, decreased GFAP, and increased Iba-1 expressions. RIP I and RIP II reversed these alterations. These results contribute to the understanding the mechanisms underlying the antidepressant effect of RIP I and RIP II, which may be related to neuroinflammatory suppression.


Subject(s)
Antidepressive Agents , Astrocytes , Depression , Disease Models, Animal , Hippocampus , Microglia , Neuroinflammatory Diseases , Stress, Psychological , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Mice , Male , Microglia/drug effects , Microglia/metabolism , Antidepressive Agents/pharmacology , Depression/drug therapy , Depression/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/complications , Stress, Psychological/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Fluoxetine/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/metabolism , Neuroprotective Agents/pharmacology , Behavior, Animal/drug effects , Glial Fibrillary Acidic Protein/metabolism
14.
Neurochem Res ; 49(10): 2957-2971, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39088164

ABSTRACT

Depression and anxiety disorders are prevalent stress-related neuropsychiatric disorders and involve multiple molecular changes and dysfunctions across various brain regions. However, the specific and shared pathophysiological mechanisms occurring in these regions remain unclear. Previous research used a rat model of chronic mild stress (CMS) to segregate and identify depression-susceptible, anxiety-susceptible, and insusceptible groups; then the proteomes of six distinct brain regions (the hippocampus, prefrontal cortex, hypothalamus, pituitary, olfactory bulb, and striatum) were separately and quantitatively analyzed. To gain a comprehensive and systematic understanding of the molecular abnormalities, this study aimed to investigate and compare differential proteomics data from the six regions. Differentially expressed proteins (DEPs) were identified in between specific regions and across all regions and subjected to a series of bioinformatics analyses. Regional comparisons showed that stress-induced proteomic changes and corresponding gene ontology and pathway enrichments were largely distinct, attributable to differences in cell populations, protein compositions, and brain functions of these areas. Additionally, a notable degree of overlap in the significantly enriched terms was identified, potentially suggesting strong connections in the enrichment across different regions. Furthermore, intra-regional and inter-regional protein-protein interaction networks and drug-target-DEP networks were constructed. Integrated analysis of the three association networks in the six regions, along with the DisGeNET database, identified ten DEPs as potential targets for anti-depression/anxiety drugs. Collectively, these findings revealed commonalities and differences across different brain regions at the protein level induced by CMS, and identified several novel protein targets for the development of new therapeutics for depression and anxiety.


Subject(s)
Anti-Anxiety Agents , Brain , Proteome , Rats, Sprague-Dawley , Stress, Psychological , Animals , Stress, Psychological/metabolism , Stress, Psychological/drug therapy , Proteome/metabolism , Male , Brain/metabolism , Brain/drug effects , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/metabolism , Anxiety/drug therapy , Depression/metabolism , Depression/drug therapy , Protein Interaction Maps , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Rats , Proteomics
15.
Molecules ; 29(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125051

ABSTRACT

Corticotropin-releasing factor (CRF) is a key neuropeptide hormone that is secreted from the hypothalamus. It is the master hormone of the HPA axis, which orchestrates the physiological and behavioral responses to stress. Many disorders, including anxiety, depression, addiction relapse, and others, are related to over-activation of this system. Thus, new molecules that may interfere with CRF receptor binding may be of value to treat neuropsychiatric stress-related disorders. Also, CRF1R antagonists have recently emerged as potential treatment options for congenital adrenal hyperplasia. Previously, several series of CRF1 receptor antagonists were developed by our group. In continuation of our efforts in this direction, herein we report the synthesis and biological evaluation of a new series of CRF1R antagonists. Representative compounds were evaluated for their binding affinities compared to antalarmin. Four compounds (2, 5, 20, and 21) showed log IC50 values of -8.22, -7.95, -8.04, and -7.88, respectively, compared to -7.78 for antalarmin. This result indicates that these four compounds are superior to antalarmin by 2.5, 1.4, 1.7, and 1.25 times, respectively. It is worth mentioning that compound 2, in terms of IC50, is among the best CRF1R antagonists ever developed in the last 40 years. The in silico physicochemical properties of the lead compounds showed good drug-like properties. Thus, further research in this direction may lead to better and safer CRF receptor antagonists that may have clinical applications, particularly for stress-related disorders and the treatment of congenital adrenal hyperplasia.


Subject(s)
Adrenal Hyperplasia, Congenital , Drug Design , Pyrimidines , Receptors, Corticotropin-Releasing Hormone , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/chemical synthesis , Humans , Adrenal Hyperplasia, Congenital/drug therapy , Adrenal Hyperplasia, Congenital/metabolism , Pyrroles/chemistry , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/drug therapy , Molecular Docking Simulation
16.
Brain Res ; 1844: 149196, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39181223

ABSTRACT

This work was done to investigate the ameliorating impact of 4-methylumbilliferon (4-MU) on spatial learning and memory dysfunction and restraint stress (STR)-induced anxiety-like behaviors in male Wistar rats and the underlying mechanisms. Thirty-two animals were assigned into 4 cohorts: control, 4-MU, STR, and STR+4-MU. Animals were exposed to STR for 4 h per day for 14 consecutive days or kept in normal conditions (healthy animals without exposure to stress). 4-MU (25 mg/kg) was intraperitoneally administered once daily to STR rats before restraint stress for 14 consecutive days. The behavioral tests were performed through Morris water maze tests and elevated-plus maze to examine learning/memory function, and anxiety levels, respectively. The levels of the antioxidant defense biomarkers (GPX, SOD) and MDA as an oxidant molecule in the brain tissues were measured using commercial ELISA kits. Neuronal loss or density of neurons was evaluated using Nissl staining. STR exposure could cause significant alterations in the levels of the antioxidant defense biomarkers (MDA, GPX, and SOD) in the prefrontal cortex and hippocampus, induce anxiety, and impair spatial learning and memory function. Treatment with 4-MU markedly reduced anxiety levels and improved spatial learning and memory dysfunction via restoring the antioxidant defense biomarkers to normal values and reducing MDA levels. Moreover, more intact cells with normal morphologies were detected in STR-induced animals treated with 4-MU. 4-MU could attenuate the STR-induced anxiety-like behaviors and spatial learning and memory dysfunction by reducing oxidative damage and neuronal loss in the prefrontal cortex and hippocampus region. Taken together, our findings provide new insights regarding the potential therapeutic effects of 4-MU against neurobehavioral disorders induced by STR.


Subject(s)
Anxiety , Cell Death , Memory Disorders , Neurons , Oxidative Stress , Rats, Wistar , Animals , Oxidative Stress/drug effects , Male , Anxiety/drug therapy , Anxiety/metabolism , Memory Disorders/drug therapy , Memory Disorders/metabolism , Neurons/drug effects , Neurons/metabolism , Rats , Cell Death/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Stress, Psychological/complications , Maze Learning/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Antioxidants/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
17.
Behav Brain Res ; 475: 115223, 2024 Oct 18.
Article in English | MEDLINE | ID: mdl-39214423

ABSTRACT

Excessive stress can exceed the adjustment ability of body and cause injury and dysfunction, while elucidation of the mechanism and prevention measures of stress-related injury are still insufficient. The present study was to observe the effect of glucocorticoid (GC) on stress-induced hypothalamic nerve injury and elucidate the potential mechanism. The present study intended to establish a chronic restraint stress rat model for follow-up study. Open field test and elevated plus maze test were used to observe behavioral changes of stress rats; Enzyme-linked immunosorbent assay (ELISA) was used to detect changes in the levels of hypothalamus-pituitary-adrenal (HPA) axis-related hormones and inflammatory factors in hypothalamus; toluidine blue staining was used to observe pathological changes of hypothalamus. The results showed that stress rats showed obvious anxiety-like behaviors, the levels of HPA axis-related hormones and inflammatory factors showed abnormal fluctuations, and morphological results showed significant nerve injury in hypothalamus. Low-dose GC treatment significantly improved behavioral changes, alleviated hypothalamic nerve injury, and restored hypothalamic levels of inflammatory factors, serum levels of GC, corticotropin-releasing hormone (CRH), and adrenocorticotropic hormone (ACTH) and GC level in adrenal cortex of stressed rats, while GC receptor (GR) inhibitor, CRH receptor inhibitor, and adrenalectomy reversed the ameliorative effects of low-dose GC. Our study clarified that low-dose GC can restore stress coping ability by reshaping the homeostasis of the HPA axis, thus alleviating behavioral abnormalities and hypothalamic nerve injury in stressed rats.


Subject(s)
Adrenocorticotropic Hormone , Glucocorticoids , Homeostasis , Hypothalamo-Hypophyseal System , Hypothalamus , Pituitary-Adrenal System , Rats, Sprague-Dawley , Stress, Psychological , Animals , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Male , Rats , Glucocorticoids/pharmacology , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Homeostasis/drug effects , Homeostasis/physiology , Adrenocorticotropic Hormone/blood , Hypothalamus/drug effects , Hypothalamus/metabolism , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Disease Models, Animal , Behavior, Animal/drug effects
18.
Neuropharmacology ; 259: 110118, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39153731

ABSTRACT

The decline of microglia in the dentate gyrus is a new phenomenon that may explain the pathogenesis of depression, and reversing this decline has an antidepressant effect. The development of strategies that restore the function of dentate gyrus microglia in under stressful conditions is becoming a new focus. Lymphocyte-activating gene-3 (LAG3) is an immune checkpoint expressed by immune cells including microglia. One of its functions is to suppress the expansion of immune cells. In a recent study, chronic systemic administration of a LAG3 antibody that readily penetrates the brain was reported to reverse chronic stress-induced hippocampal microglia decline and depression-like behaviors. We showed here that a single intranasal infusion of a LAG3 antibody (In-LAG3 Ab) reversed chronic unpredictable stress (CUS)-induced depression-like behaviors in a dose-dependent manner, which was accompanied by an increase in brain-derived neurotrophic factor (BDNF) in the dentate gyrus. Infusion of an anti-BDNF antibody into the dentate gyrus, construction of knock-in mice with the BDNF Val68Met allele, or treatment with the BDNF receptor antagonist K252a abolished the antidepressant effect of In-LAG3 Ab. Activation of extracellular signal-regulated kinase1/2 (ERK1/2) is required for the reversal effect of In-LAG3 Ab on CUS-induced depression-like behaviors and BDNF decrease in the dentate gyrus. Moreover, both inhibition and depletion of microglia prevented the reversal effect of In-LAG3 Ab on CUS-induced depression-like behaviors and impairment of ERK1/2-BDNF signaling in the dentate gyrus. These results suggest that In-LAG3 Ab exhibits an antidepressant effect through microglia-mediated activation of ERK1/2 and synthesis of BDNF in the dentate gyrus.


Subject(s)
Administration, Intranasal , Antidepressive Agents , Antigens, CD , Brain-Derived Neurotrophic Factor , Depression , Hippocampus , Lymphocyte Activation Gene 3 Protein , MAP Kinase Signaling System , Stress, Psychological , Animals , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Male , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Mice , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Depression/drug therapy , Antigens, CD/metabolism , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Antibodies/pharmacology , Carbazoles/pharmacology , Carbazoles/administration & dosage , Signal Transduction/drug effects , Indole Alkaloids
19.
Eur J Pharmacol ; 980: 176869, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39117265

ABSTRACT

Depressive pseudodementia (DPD) is a debilitating cognitive dysfunction that accompanies major and/or frequent depressive attacks. DPD has gained significant research attention owing to its negative effects on the patients' quality of life and productivity. This study tested the procognitive potential of Flibanserin (FBN), the serotonin (5HT) receptor modulator, against propranolol (PRP), as ß/5HT1A receptors blocker. Serving this purpose, female Wistar Albino rats were subjected to chronic unpredictable stress (CUS) and subsequently treated with FBN only (3 mg/kg/day, p.o), PRP only (10 mg/kg/day, p.o), or PRP followed by FBN, using the same doses. FBN ameliorated the behavioral/cognitive alterations and calmed the hypothalamic-pituitary-adrenal (HPA) axis storm by reducing the levels of stress-related hormones, viz, corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), corticosterone (CORT) parallel to epinephrine (EPI) hyperstimulation. The maladaptive inflammatory response, comprising of interleukin (IL)-1ß/6, and tumor necrosis factor (TNF)-α, was consequently blunted. This was contemporaneous to the partial restoration of the protein kinase-B (AKT)/glycogen synthase kinase (GSK)3ß/signal transducer and activator of transcription (STAT)-3 survival trajectory and the reinstatement of the levels of brain derived neurotrophic factor (BDNF). Microscopically, FBN repaired the hippocampal architecture and lessened CD68/GFAP immunoreactivity. Pre-administration of PRP partially abolished FBN effect along the estimated parameters, except for 5HT2A receptor expression and epinephrine level, to prove 5HT1A receptor as a fulcrum initiator of the investigated pathway, while its sole administration worsened the underlying condition. Ultimately, these findings highlight the immense procognitive potential of FBN, offering a new paradigm for halting DPD advancement via synchronizing adrenergic/serotonergic circuitry.


Subject(s)
Benzimidazoles , Brain-Derived Neurotrophic Factor , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Proto-Oncogene Proteins c-akt , Rats, Wistar , Animals , Female , Rats , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Serotonin/metabolism , Signal Transduction/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/complications , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use
20.
Eur J Pharmacol ; 982: 176939, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39182548

ABSTRACT

The efficacy and tolerability of current antidepressants for adolescent depression are inadequate. S-adenosylmethionine (SAMe), known for its effectiveness and minimal side effects in adult depression, remains unstudied in adolescents. This study explored the potential of SAMe to address depression-like behaviors in juvenile rats induced by chronic unpredictable mild stress (CUMS), with a focus on gut microbiome interactions. Adolescent male Wistar rats were subjected to a 4-week CUMS regimen and received daily intraperitoneal injections of 300 mg/kg SAMe. Behavioral assessments included the sucrose preference test, elevated plus maze test, open field test, and Y-maze test. Histopathological changes of the hippocampus and colon were observed by Nissl staining and hematoxylin and eosin staining, respectively. Gut microbiome composition was analyzed using Accurate 16S absolute quantification sequencing. The results showed that SAMe significantly improved behavioral outcomes, reduced histopathological damages in hippocampal neurons and colon tissues, and modulated the gut microbiota of depressed rats. It favorably altered the ratio of Bacteroidetes to Firmicutes, decreased the absolute abundance of Deferribacteres, and adjusted levels of key microbial genera associated with depression-like behaviors. These results suggested that SAMe could effectively counter depression-like behaviors in CUMS-exposed adolescent rats by mitigating hippocampal neuronal and colon damage and modulating the gut microbiota. This supports SAMe as a viable and tolerable treatment option for adolescent depression, highlighting the importance of the gut-brain axis in therapeutic strategies.


Subject(s)
Behavior, Animal , Depression , Gastrointestinal Microbiome , Hippocampus , Rats, Wistar , S-Adenosylmethionine , Stress, Psychological , Animals , Gastrointestinal Microbiome/drug effects , Male , Depression/drug therapy , Stress, Psychological/complications , Stress, Psychological/drug therapy , S-Adenosylmethionine/pharmacology , S-Adenosylmethionine/therapeutic use , Hippocampus/drug effects , Hippocampus/pathology , Rats , Behavior, Animal/drug effects , Colon/drug effects , Colon/pathology , Colon/microbiology , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL