Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.762
Filter
1.
J Ethnopharmacol ; 336: 118739, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39197805

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Modified Danzhi Xiaoyao San (MDXS) is an effective clinical prescription for depression in China, which was deprived of Danzhi Xiaoyao San in the Ming Dynasty. MDSX has significant implications for the development of new antidepressants, but its pharmacological mechanism has been rarely studied. AIM OF THE STUDY: To reveal the active components and molecular mechanism of MDXS in treating depression through network pharmacology and experimental verification in vivo and in vitro. MATERIALS AND METHODS: UPLC-Q-TOF-MS/MS was used to identify the chemical components in the MDXS freeze-dried powder, drug-containing serum, and cerebrospinal fluid (CSF). Based on the analysis of prototype components in the CSF, the major constituents, potential therapeutic targets and possible pharmacological mechanisms of MDXS in treating depression were investigated using network pharmacological and molecular docking. Then corticosterone (CORT)-induced mice model of depression was established to investigate the antidepressant effects of MDXS. HT22 cells were cultured to verify the neuroprotective effects and core targets of the active components. RESULTS: There were 81 compounds in MDXS freeze-dried powder, 36 prototype components in serum, and 13 prototype components in CSF were identified, respectively. Network pharmacology analysis showed that these 13 prototype components in the CSF shared 190 common targets with depression, which were mainly enriched in MAPK and PI3K/AKT signaling pathways. PPI analysis suggested that AKT1 and MAPK1 (ERK1/2) were the core targets. Molecular docking revealed that azelaic acid (AA), senkyunolide A (SA), atractylenolide III (ATIII), and tokinolide B (TB) had the highest binding energy with AKT1 and MAPK1. Animal experiments verified that MDXS could reverse CORT-induced depression-like behaviors, improve synaptic plasticity, alleviate neuronal injury in hippocampal CA3 regions, and up-regulate the protein expression of p-ERK1/2 and p-AKT. In HT22 cells, azelaic acid, senkyunolide A, and atractylenolide III significantly protected the cell injury caused by CORT, and up-regulated the protein levels of p-ERK1/2 and p-AKT. CONCLUSIONS: These results suggested that MDXS may exert antidepressant effects partially through azelaic acid, senkyunolide A, and atractylenolide III targeting ERK1/2 and AKT.


Subject(s)
Antidepressive Agents , Depression , Drugs, Chinese Herbal , Molecular Docking Simulation , Network Pharmacology , Animals , Antidepressive Agents/pharmacology , Depression/drug therapy , Drugs, Chinese Herbal/pharmacology , Mice , Male , Cell Line , Disease Models, Animal , Mice, Inbred C57BL , Corticosterone/blood , Tandem Mass Spectrometry , Behavior, Animal/drug effects
2.
Yale J Biol Med ; 97(3): 365-381, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39351321

ABSTRACT

Depression is a significant mental health challenge globally. While traditional antidepressants are effective, they often have unwanted side effects. Saffron, a natural spice derived from Crocus sativus L., has emerged as a potential alternative therapy for depression. Researchers have found that its components such as crocin, crocetin, and safranal have been found to mitigate depressive symptoms through neurotransmitter regulation, anti-inflammatory effects, and neuroprotection. Clinical trials suggest that the effectiveness of saffron in treating mild to moderate depression is comparable to that of standard medications, and animal studies support these results, showing behavioral improvements with saffron treatment. Saffron is particularly appealing due to its safety and lower incidence of side effects, making it suitable for those sensitive to conventional drugs. Additionally, its antioxidant properties may offer further health benefits. However, challenges such as determining the appropriate dosage, prohibitive cost, and the limited availability of quality saffron need to be addressed. Most research on saffron's efficacy is short-term; thus, long-term studies are essential to understand its full therapeutic potential and ongoing antidepressant effects. While saffron is safe in terms of its culinary value, higher therapeutic doses require careful monitoring for drug interactions and side effects. In summary, saffron represents a promising direction in depression treatment, with benefits potentially matching those of standard treatments and a better safety profile. However, further research is necessary to establish clear guidelines for its use, optimize dosing, and assess long-term outcomes. Saffron offers a natural treatment path for depression, but its use must be controlled and supported by scientific evidence.


Subject(s)
Antidepressive Agents , Crocus , Depression , Crocus/chemistry , Humans , Depression/drug therapy , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Animals , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
3.
Transl Psychiatry ; 14(1): 400, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358323

ABSTRACT

Major depressive disorder (MDD) is a common disease affecting 300 million people worldwide. The existing drugs are ineffective for approximately 30% of patients, so it is urgent to develop new antidepressant drugs with novel mechanisms. Here, we found that norisoboldine (NOR) showed an antidepressant efficacy in the chronic social defeat stress (CSDS) depression model in the tail suspension, forced swimming, and sucrose consumption tests. We then utilized the drug-treated CSDS mice paradigm to segregate and gain differential protein groups of CSDS versus CON (CSDSCON), imipramine (IMI)-treated versus CSDS (IMICSDS), and NOR-treated versus CSDS (NORCSDS) from the prefrontal cortex. These protein expression alterations were first analyzed by ANOVA with p < 0.05. The protein cluster 1 and cluster 3, in which the pattern of protein levels similar to the mood pattern, showed enrichment in functions and localizations related to mitochondrion, ribosome and synapses. Further GO analysis of the common proteins for NORCSDS groups and NORIMI groups supported the findings from ANOVA analysis. We employed Protein-Protein interaction (PPI) analysis to examine the proteins of NORCSDS and NORIMI, revealing an enrichment of the proteins associated with the mitochondrial ribosomal and synaptic functions. Further independent analysis using parallel reaction monitoring (PRM) revealed that Cox7c, Mrp142, Naa30, Ighm, Apoa4, Ssu72, Mrps30, Apoh, Acbd5, and Cdv3, exhibited regulation in the NOR-treated group to support the homeostasis of mitochondrial functions. Additionally, Dcx, Arid1b, Rnf112, and Fam3c, were also observed to undergo modulation in the NOR-treated groups to support the synaptic formation and functions. These findings suggest that the proteins involved in depression treatment exert effects in strengthen the mitochondrial and synaptic functions in the mice PFC. Western blot analysis supported the data that the levels of Mrpl42, Cox7c, Naa30, Rnf112, Dcx Apoa4, Apoh and Fam3c were altered in the CSDS mice, and rescued by NOR treatment, supporting the PRM data. NOR treatment also rescued the NLRP3 inflammasome activation in CSDS mice. In summary, the current proteomic research conducted on the prefrontal cortex has provided valuable insights into the specific and shared molecular mechanisms underlying pathophysiology and treatment to CSDS-induced depression, shedding light on the therapeutic effects of Norisoboldine.


Subject(s)
Antidepressive Agents , Disease Models, Animal , Mitochondria , Prefrontal Cortex , Proteomics , Stress, Psychological , Animals , Mice , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Male , Mitochondria/drug effects , Mitochondria/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Synapses/drug effects , Synapses/metabolism , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Mice, Inbred C57BL , Doublecortin Protein , Depression/drug therapy , Depression/metabolism , Behavior, Animal/drug effects , Social Defeat
4.
Transl Psychiatry ; 14(1): 411, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358318

ABSTRACT

Depression is a common and complex psychiatric illness with multiple clinical symptoms, even leading to the disability and suicide. Owing to the partial understanding of the pathogenesis of depressive-like disorders, available pharmacotherapeutic strategies are developed mainly based on the "monoamine hypothesis", resulting in a limited effectiveness and a number of adverse effects in the clinical practice. The concept of multiple pathogenic factors be helpful for clarifying the etiology of depression and developing the antidepressants. It is well documented that K+ channels serve crucial roles in modulating the neuronal excitability and neurotransmitter release in the brain, and abnormality of these channels participated in the pathogenic process of diverse central nervous system (CNS) pathologies, such as seizure and Alzheimer's disease (AD). The clinical and preclinical evidence also delineates that the involvement of several types of K+ channels in depressive-like behaviors appear to be evident, suggesting these channels being one of the multiple factors in the etiology of this debilitating disorder. Emerging data manifest that diverse antidepressants impact distinct K+ channels, such as Kv, Kir and K2P, meaning the functioning of these drug via a "multi-target" manner. On the other hand, the scenario of antidepressants impinging K+ channels could render an alternative interpretation for the pharmacological effectiveness and numerous side effects in clinical trials. Furthermore, these channels serve to be considered as a "druggable target" to develop novel therapeutic compound to antagonize this psychiatry.


Subject(s)
Antidepressive Agents , Potassium Channels , Humans , Antidepressive Agents/pharmacology , Potassium Channels/metabolism , Potassium Channels/drug effects , Animals , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Depression/drug therapy , Depression/metabolism , Brain/drug effects , Brain/metabolism
5.
Biol Pharm Bull ; 47(10): 1637-1643, 2024.
Article in English | MEDLINE | ID: mdl-39370268

ABSTRACT

Depressive disorder is the most common mental disorder with significant economic burden and limited treatments. Traditional Chinese medicine monomer has emerged as a promising non-pharmacological treatment for reducing depressive symptoms. The aim of this study was to investigate the antidepressant-like effects of asperuloside (ASP) and its mechanism. The depression-like behaviors of chronic unpredictable mild stress (CUMS)-exposed rats were evaluated by behavioral tests. At the same time, the behaviors of rats treated with different concentrations of ASP (10, 20, 40 mg/kg) were also evaluated. RNA sequencing was performed to screen for dysregulated genes following ASP treatment. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was performed to state the enriched pathways. Protein expression was detected by Western blotting. With the increase of ASP concentration (over 20 mg/kg), the depression-like behaviors of the rats were alleviated, which was manifested as the increase of the number of entries in the central zone, decrease of immobility time, and the increase of swimming time, sucrose preference, and body weight. ASP activated the Wnt3α/glycogen synthase kinase 3ß (GSK-3ß)/ß-catenin signaling pathway in vivo. Knockdown of ß-catenin reversed the effects of ASP on regulating depression-like behaviors. ASP alleviates depression-like behaviors by activating the Wnt3α/GSK-3ß/ß-catenin signaling pathway, indicating that ASP may be a potential therapeutic drug for treatment of depression.


Subject(s)
Antidepressive Agents , Depression , Glycogen Synthase Kinase 3 beta , Rats, Sprague-Dawley , Animals , Glycogen Synthase Kinase 3 beta/metabolism , Male , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depression/drug therapy , Wnt3 Protein/metabolism , Wnt3 Protein/genetics , beta Catenin/metabolism , Behavior, Animal/drug effects , Rats , Signal Transduction/drug effects , Stress, Psychological/drug therapy , Hippocampus/metabolism , Hippocampus/drug effects
6.
J Pharmacol Sci ; 156(3): 180-187, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39313276

ABSTRACT

Fibromyalgia (FM) is an intractable disease with a chief complaint of chronic widespread pain. Amitriptyline (AMI) and duloxetine (DLX), which are antidepressant drugs, have been reported to ameliorate pain in patients with FM and pain-related behaviors in several rodent models of FM. However, the mechanisms of action of AMI and DLX are not yet fully understood. Here, we examined the effects of these drugs on the responsiveness of superficial dorsal horn (SDH) neurons in the spinal cord, using a rat FM model developed by injecting a biogenic amine depleter (reserpine). Extracellular recordings of SDH neurons in vivo demonstrated that bath application of AMI and DLX at concentrations of 0.1-1.0 mM on the dorsal surface of the spinal cord markedly suppressed spontaneous discharge and von Frey filament-evoked mechanical firing in SDH neurons. The suppression induced by the drugs was noted in a concentration-dependent manner and the suppressive effects resolved after washing the spinal cord surface. These results show that SDH neurons are the site of action for AMI and DLX in a rat reserpine-induced FM model. Spinal mechanisms may underlie the therapeutic effects of these drugs in patients with FM.


Subject(s)
Amitriptyline , Disease Models, Animal , Duloxetine Hydrochloride , Fibromyalgia , Posterior Horn Cells , Rats, Sprague-Dawley , Reserpine , Animals , Duloxetine Hydrochloride/pharmacology , Amitriptyline/pharmacology , Fibromyalgia/drug therapy , Fibromyalgia/chemically induced , Posterior Horn Cells/drug effects , Male , Rats , Antidepressive Agents/pharmacology , Dose-Response Relationship, Drug
7.
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
8.
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
9.
Phytomedicine ; 134: 156012, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39260135

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is a severe mental health condition characterized by persistent depression, impaired cognition, and reduced activity. Increasing evidence suggests that gut microbiota (GM) imbalance is closely linked to the emergence and advancement of MDD, highlighting the potential significance of regulating the "Microbiota-Gut-Brain" (MGB) axis to impact the development of MDD. Natural products (NPs), characterized by broad biological activities, low toxicity, and multi-target characteristics, offer unique advantages in antidepressant treatment by regulating MGB axis. PURPOSE: This review was aimed to explore the intricate relationship between the GM and the brain, as well as host responses, and investigated the mechanisms underlying the MGB axis in MDD development. It also explored the pharmacological mechanisms by which NPs modulate MGB axis to exert antidepressant effects and addressed current research limitations. Additionally, it proposed new strategies for future preclinical and clinical applications in the MDD domain. METHODS: To study the effects and mechanism by which NPs exert antidepressant effects through mediating the MGB axis, data were collected from Web of Science, PubMed, ScienceDirect from initial establishment to March 2024. NPs were classified and summarized by their mechanisms of action. RESULTS: NPs, such as flavonoids,alkaloids,polysaccharides,saponins, terpenoids, can treat MDD by regulating the MGB axis. Its mechanism includes balancing GM, regulating metabolites and neurotransmitters such as SCAFs, 5-HT, BDNF, inhibiting neuroinflammation, improving neural plasticity, and increasing neurogenesis. CONCLUSIONS: NPs display good antidepressant effects, and have potential value for clinical application in the prevention and treatment of MDD by regulating the MGB axis. However, in-depth study of the mechanisms by which antidepressant medications affect MGB axis will also require considerable effort in clinical and preclinical research, which is essential for the development of effective antidepressant treatments.


Subject(s)
Antidepressive Agents , Biological Products , Brain-Gut Axis , Depressive Disorder, Major , Gastrointestinal Microbiome , Antidepressive Agents/pharmacology , Humans , Gastrointestinal Microbiome/drug effects , Brain-Gut Axis/drug effects , Brain-Gut Axis/physiology , Depressive Disorder, Major/drug therapy , Biological Products/pharmacology , Animals , Brain/drug effects
10.
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
11.
Adv Exp Med Biol ; 1456: 49-66, 2024.
Article in English | MEDLINE | ID: mdl-39261423

ABSTRACT

Major depressive disorder (MDD) is a highly prevalent psychiatric disorder, associated with substantial burden and large economical costs. Notwithstanding various conventional antidepressant treatment options, a large portion of depressed people (ca. 30%) fails to respond to first-line treatment, resulting in treatment-resistant depression (TRD). Although non-response to multiple antidepressant interventions is a common outcome, a consensus definition of TRD is not yet available. In practice, TRD is applied when two or more successive treatments with different antidepressants are not working. The last decade's intense research into new medicines for TRD has led to two developments, using typical or serotonergic (psilocybin, ayahuasca) and atypical (glutamatergic) psychedelics (ketamine, esketamine). Both approaches, although via different entrance mechanism, exhibit a fast onset but also long-lasting antidepressant effect far beyond the biological presence of the drug in the body, strongly indicating that downstream mechanisms activated by signaling cascades in the brain are involved. The present chapter describes the clinical development of psilocybin and esketamine for TRD and discusses the problems involved in the use of a proper placebo because of the psychotomimetic (psilocybin) or dissociative (ketamine) effects that interfere with performing "blind" studies. Nevertheless, intranasal esketamine was developed and approved for TRD, whereas psilocybin has shown positive results. Adverse effects and tolerability of both drugs in the dose ranges used are generally acceptable. The emergence of anti-TRD medicines for treatment of a very severe disease is a breakthrough in psychiatry.


Subject(s)
Antidepressive Agents , Depressive Disorder, Treatment-Resistant , Hallucinogens , Ketamine , Psilocybin , Humans , Depressive Disorder, Treatment-Resistant/drug therapy , Hallucinogens/therapeutic use , Hallucinogens/adverse effects , Hallucinogens/pharmacology , Ketamine/therapeutic use , Ketamine/adverse effects , Psilocybin/therapeutic use , Psilocybin/adverse effects , Psilocybin/pharmacology , Antidepressive Agents/therapeutic use , Antidepressive Agents/adverse effects , Antidepressive Agents/pharmacology , Depressive Disorder, Major/drug therapy , Treatment Outcome
12.
Adv Exp Med Biol ; 1456: 27-48, 2024.
Article in English | MEDLINE | ID: mdl-39261422

ABSTRACT

Major depressive disorder (MDD) is a mental health disorder associated with cognitive impairment, dysregulated appetite, fatigue, insomnia or hypersomnia, and severe mood changes that significantly impact the ability of the affected individual to perform day-to-day tasks, leading to suicide in the worst-case scenario. As MDD is becoming more prevalent, affecting roughly 300 million individuals worldwide, its treatment has become a major point of interest. Antidepressants acting as selective serotonin reuptake inhibitors (SSRIs) are currently used as the first line of treatment for MDD. Other antidepressants currently used for the treatment of MDD include the serotonin and norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), and monoamine oxidase inhibitors (MAOIs). However, although effective in alleviating symptoms of MDD, most antidepressants require weeks or even months of regular administration prior to eliciting a rational clinical effect. Owing to the strong evidence showing a relationship between neural plasticity, neurogenesis, and MDD, researchers have also looked at the possibility of using treatment modalities that target these processes in an attempt to improve clinical outcome. The overarching aim of this chapter is to highlight the role of neural plasticity and neurogenesis in the pathophysiology of MDD and discuss the most recently studied treatment strategies that target these processes by presenting supporting evidence from both animal and human studies.


Subject(s)
Antidepressive Agents , Depressive Disorder, Major , Neurogenesis , Neuronal Plasticity , Humans , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/physiopathology , Neurogenesis/drug effects , Neuronal Plasticity/drug effects , Antidepressive Agents/therapeutic use , Antidepressive Agents/pharmacology , Animals , Selective Serotonin Reuptake Inhibitors/therapeutic use , Selective Serotonin Reuptake Inhibitors/pharmacology
13.
Rev Med Suisse ; 20(885): 1540-1543, 2024 Sep 04.
Article in French | MEDLINE | ID: mdl-39238456

ABSTRACT

The mechanism of action of selective serotonin reuptake inhibitors (SSRI) is still not properly established. It is essential to consider their positive and negative side effects before prescribing. In this article, we describe several of these side effects in the context of common pathologies and clinical situations. We discuss their cardioprotective effect and their role in the functional recovery of patients following stroke. We recall the increase in the risk of bleeding when prescribing SSRI concomitantly with antiaggregating and anticoagulant treatments. Prescribing SSRI also increases the risk of fracture and the frequency of hyponatremia. In the context of COPD, the effects of SSRI are more difficult to establish.


Le mécanisme d'action des antidépresseurs inhibiteurs sélectifs de la recapture de la sérotonine (ISRS) n'est toujours pas formellement établi. Il est essentiel de prendre en compte leurs effets secondaires positifs et négatifs pour leur prescription. Dans cet article, nous décrivons plusieurs de ces effets dans le contexte de pathologies et situations cliniques courantes. Nous abordons leur effet cardioprotecteur ainsi que leur rôle dans la récupération fonctionnelle des patients à la suite des accidents vasculaires cérébraux. Nous rappelons la majoration du risque hémorragique lors de la prescription d'ISRS en concomitance de traitements antiagrégants et anticoagulants. La prescription d'ISRS augmente également le risque fracturaire et la fréquence d'une hyponatrémie. Dans le contexte de la bronchopneumopathie chronique obstructive, les effets d'un ISRS sont plus difficiles à établir.


Subject(s)
Selective Serotonin Reuptake Inhibitors , Humans , Selective Serotonin Reuptake Inhibitors/adverse effects , Antidepressive Agents/adverse effects , Antidepressive Agents/pharmacology , Stroke/prevention & control , Stroke/chemically induced , Hemorrhage/chemically induced , Fractures, Bone/prevention & control , Fractures, Bone/chemically induced
14.
Transl Psychiatry ; 14(1): 359, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231927

ABSTRACT

BACKGROUND: Rapid-acting antidepressants (RAADs), including dissociative anesthetics, psychedelics, and empathogens, elicit rapid and sustained therapeutic improvements in psychiatric disorders by purportedly modulating neuroplasticity, neurotransmission, and immunity. These outcomes may be mediated by, or result in, an acute and/or sustained entrainment of epigenetic processes, which remodel chromatin structure and alter DNA accessibility to regulate gene expression. METHODS: In this perspective, we present an overview of the known mechanisms, knowledge gaps, and future directions surrounding the epigenetic effects of RAADs, with a focus on the regulation of stress-responsive DNA and brain regions, and on the comparison with conventional antidepressants. MAIN BODY: Preliminary correlative evidence indicates that administration of RAADs is accompanied by epigenetic effects which are similar to those elicited by conventional antidepressants. These include changes in DNA methylation, post-translational modifications of histones, and differential regulation of non-coding RNAs in stress-responsive chromatin areas involved in neurotrophism, neurotransmission, and immunomodulation, in stress-responsive brain regions. Whether these epigenetic changes causally contribute to the therapeutic effects of RAADs, are a consequence thereof, or are unrelated, remains unknown. Moreover, the potential cell type-specificity and mechanisms involved are yet to be fully elucidated. Candidate mechanisms include neuronal activity- and serotonin and Tropomyosine Receptor Kinase B (TRKB) signaling-mediated epigenetic changes, and direct interaction with DNA, histones, or chromatin remodeling complexes. CONCLUSION: Correlative evidence suggests that epigenetic changes induced by RAADs accompany therapeutic and side effects, although causation, mechanisms, and cell type-specificity remain largely unknown. Addressing these research gaps may lead to the development of novel neuroepigenetics-based precision therapeutics.


Subject(s)
Antidepressive Agents , DNA Methylation , Epigenesis, Genetic , Epigenesis, Genetic/drug effects , Humans , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , DNA Methylation/drug effects , Brain/drug effects , Brain/metabolism , Animals , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Histones/metabolism , Stress, Psychological/genetics
15.
Int J Mol Sci ; 25(17)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39273621

ABSTRACT

Depression is a prevalent and debilitating mental disorder that affects millions worldwide. Current treatments, such as antidepressants targeting the serotonergic system, have limitations, including delayed onset of action and high rates of treatment resistance, necessitating novel therapeutic strategies. Ginsenoside Rc (G-Rc) has shown potential anti-inflammatory and neuroprotective effects, but its antidepressant properties remain unexplored. This study investigated the antidepressant effects of G-Rc in an L-alpha-aminoadipic acid (L-AAA)-induced mouse model of depression, which mimics the astrocytic pathology and neuroinflammation observed in major depressive disorder. Mice were administered G-Rc, vehicle, or imipramine orally after L-AAA injection into the prefrontal cortex. G-Rc significantly reduced the immobility time in forced swimming and tail suspension tests compared to vehicle treatment, with more pronounced effects than imipramine. It also attenuated the expression of pro-inflammatory cytokines (TNF-α, IL-6, TGF-ß, lipocalin-2) and alleviated astrocytic degeneration, as indicated by increased GFAP and decreased IBA-1 levels. Additionally, G-Rc modulated apoptosis-related proteins, decreasing caspase-3 and increasing Bcl-2 levels compared to the L-AAA-treated group. These findings suggest that G-Rc exerts antidepressant effects by regulating neuroinflammation, astrocyte-microglia crosstalk, and apoptotic pathways in the prefrontal cortex, highlighting its potential as a novel therapeutic agent for depression.


Subject(s)
2-Aminoadipic Acid , Antidepressive Agents , Astrocytes , Ginsenosides , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Mice , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Ginsenosides/pharmacology , Male , 2-Aminoadipic Acid/pharmacology , Depression/drug therapy , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Disease Models, Animal , Cytokines/metabolism , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Apoptosis/drug effects
16.
J Neuroimmune Pharmacol ; 19(1): 50, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39312021

ABSTRACT

Depression is a global psychiatric illness that imposes a substantial economic burden. Unfortunately, traditional antidepressants induce many side effects which limit patient compliance thus, exploring alternative therapies with fewer adverse effects became urgent. This study aimed to investigate the effect of trimetazidine (TMZ); a well-known anti-ischemic drug in lipopolysaccharide (LPS) mouse model of depression focusing on its ability to regulate toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) as well as nuclear factor erythroid 2 related factor 2 (Nrf2)/ heme oxygenase-1 (HO-1) signaling pathways. Male Swiss albino mice were injected with LPS (500 µg/kg, i.p) every other day alone or parallel with oral doses of either escitalopram (Esc) (10 mg/kg/day) or TMZ (20 mg/kg/day) for 14 days. Treatment with TMZ attenuated LPS-induced animals' despair with reduced immobility time inforced swimming test. TMZ also diminished LPS- induced neuro-inflammation via inhibition of TLR4/NF-κB pathway contrary to Nrf2/HO-1 cascade activation with consequent increase in reduced glutathione (GSH) and HO-1 levels whereas the pro-inflammatory cytokines; tumor necrosis factor-α (TNF-α) and interleukin (IL)-1ß were evidently reduced. Besides, TMZ replenished brain serotonin levels via serotonin transporter (SERT) inhibition. Thus, TMZ hindered LPS-induced neuro-inflammation, oxidative stress, serotonin deficiency besides its anti-apoptotic effect which was reflected by decreased caspase-3 level. Neuroprotective effects of TMZ were confirmed by the histological photomicrographs which showed prominent neuronal survival. Here we showed that TMZ is an affluent nominee for depression management via targeting TLR4/NF-κB and Nrf2/HO-1 pathways. Future research addressing TMZ-antidepressant activity in humans is mandatory to enroll it as a novel therapeutic strategy for depression.


Subject(s)
Depression , Lipopolysaccharides , NF-E2-Related Factor 2 , NF-kappa B , Toll-Like Receptor 4 , Trimetazidine , Animals , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Male , Mice , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Lipopolysaccharides/toxicity , Depression/drug therapy , Depression/chemically induced , Depression/metabolism , Trimetazidine/pharmacology , Trimetazidine/therapeutic use , Signal Transduction/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Heme Oxygenase-1/metabolism , Membrane Proteins
17.
Biomed Pharmacother ; 179: 117263, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39243431

ABSTRACT

Post-stroke depression (POSD) is a common difficulty and most predominant emotional syndrome after stroke often consequences in poor outcomes. In the present investigation, we have designed and studied the neurologically active celastrol/minocycline encapsulated with macrophages-derived exosomes functionalized PLGA nanoformulations (CMC-EXPL) to achieve enhanced anti-inflammatory behaviour and anti-depressant like activity in a Rat model of POSD. The animal model of POSD was established through stimulating process with chronic unpredictable mild stress (CUM) stimulations after procedure of middle cerebral artery occlusion (MCAO). Neuronal functions and Anti-inflammation behaviours were observed by histopathological (H&E) examination and Elisa analyses, respectively. The anti-depressive activity of the nanoformulations treated Rat models were evaluated by open-field and sucrose preference test methods. Microglial polarization was evaluated via flow-cytometry and qRT-PCR observations. The observed results exhibited that prepared nanoformulations reduced the POSD-stimulated depressive-like activities in rat models as well alleviated the neuronal damages and inflammatory responses in the cerebral hippocampus. Importantly, prepared CMC-EXPL nanoformulation effectively prevented the M1 pro-inflammatory polarization and indorsed M2 anti-inflammatory polarization, which indicates iNOS and CD86 levels significantly decreased and upsurged Arg-1 and CD206 levels. CMC-EXPL nanoformulation suggestively augmented anti-depressive activities and functional capability and also alleviated brain inflammation in POSD rats, demonstrating its therapeutic potential for POSD therapy.


Subject(s)
Depression , Disease Models, Animal , Drug Carriers , Exosomes , Macrophages , Microglia , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Sprague-Dawley , Stroke , Animals , Exosomes/metabolism , Depression/drug therapy , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Microglia/drug effects , Microglia/metabolism , Male , Rats , Macrophages/drug effects , Macrophages/metabolism , Drug Carriers/chemistry , Stroke/drug therapy , Stroke/complications , Nanoparticles/chemistry , Neuroinflammatory Diseases/drug therapy , Drug Delivery Systems/methods , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/administration & dosage , Drug Compounding
18.
Int J Neuropsychopharmacol ; 27(10)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39297528

ABSTRACT

BACKGROUND: Understanding the precise mechanisms of ketamine is crucial for replicating its rapid antidepressant effects without inducing psychomimetic changes. Here, we explore whether the antidepressant-like effects of ketamine enantiomers are underscored by protection against cytokine-induced reductions in hippocampal neurogenesis and activation of the neurotoxic kynurenine pathway in our well-established in vitro model of depression in a dish. METHODS: We used the fetal hippocampal progenitor cell line (HPC0A07/03C) to investigate ketamine's impact on cytokine-induced reductions in neurogenesis in vitro. Cells were treated with interleukin- 1beta (IL-1b) (10 ng/mL) or IL-6 (50 pg/mL), alone or in combination with ketamine enantiomers arketamine (R-ketamine, 400 nM) or esketamine (S-ketamine, 400 nM) or antidepressants sertraline (1 mM) or venlafaxine (1 mM). RESULTS: Resembling the effect of antidepressants, both ketamine enantiomers prevented IL-1b- and IL-6-induced reduction in neurogenesis and increase in apoptosis. This was mediated by inhibition of IL-1b-induced production of IL-2 and IL-13 by R-ketamine and of IL-1b-induced tumor necrosis factor-alpha by S-ketamine. Likewise, R-ketamine inhibited IL-6-induced production of IL-13, whereas S-ketamine inhibited IL-6-induced IL-1b and IL-8. Moreover, both R- and S-ketamine prevented IL-1b-induced increases in indoleamine 2,3-dioxygenase expression as well as kynurenine production, which in turn was shown to mediate the detrimental effects of IL-1b on neurogenesis and apoptosis. In contrast, neither R- nor S-ketamine prevented IL-6-induced kynurenine pathway activation. CONCLUSIONS: Results suggest that R- and S-ketamine have pro-neurogenic and anti-inflammatory properties; however, this is mediated by inhibition of the kynurenine pathway only in the context of IL-1b. Overall, this study enhances our understanding of the mechanisms underlying ketamine's antidepressant effects in the context of different inflammatory phenotypes, ultimately leading to the development of more effective, personalized therapeutic approaches for patients suffering from depression.


Subject(s)
Antidepressive Agents , Hippocampus , Ketamine , Kynurenine , Neurogenesis , Ketamine/pharmacology , Kynurenine/pharmacology , Kynurenine/metabolism , Neurogenesis/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Antidepressive Agents/pharmacology , Interleukin-1beta/metabolism , Cell Line , Interleukin-6/metabolism , Inflammation/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Apoptosis/drug effects , Venlafaxine Hydrochloride/pharmacology , Sertraline/pharmacology , Stereoisomerism
19.
Biochem Biophys Res Commun ; 733: 150696, 2024 Nov 12.
Article in English | MEDLINE | ID: mdl-39288700

ABSTRACT

Major depressive disorder (MDD) is a psychiatric disorder characterized by depressed mood, behavioral despair and anhedonia. Demyelination in specific brain regions underlies the pathology of MDD, raising the alleviating demyelination as a potential strategy for MDD therapy. Nervonic acid (NA) has the potential to improve brain demyelination, offering benefits for various neurological disorders. However, its effects on depression remain undetermined. Mice were subjected to 14 days of chronic restraint stress (CRS) to induce depression-like behaviors, and were injected with NA (70 mg/kg) daily. The administration of NA significantly improved depressive-like behaviors in CRS mice. CRS led to significant demyelination in the medial prefrontal cortex (mPFC), which were reversed by NA treatment. In addition, NA ameliorated the upregulation of inflammatory cytokines and downregulation of brain-derived neurotrophic factor, improved the alternations in axonal spines observed in the mPFC of CRS mice. Our results highlighted the potential of NA as an antidepressant, with its benefits likely attributed to its effects in alleviating demyelination in the mPFC.


Subject(s)
Antidepressive Agents , Demyelinating Diseases , Depression , Mice, Inbred C57BL , Prefrontal Cortex , Restraint, Physical , Stress, Psychological , Animals , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Male , Stress, Psychological/drug therapy , Mice , Demyelinating Diseases/drug therapy , Demyelinating Diseases/pathology , Depression/drug therapy , Depression/pathology , Depression/metabolism , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism
20.
Sci Rep ; 14(1): 22111, 2024 09 27.
Article in English | MEDLINE | ID: mdl-39333605

ABSTRACT

Lactoferrin is a natural multifunctional glycoprotein with potential antidepressant-like effects. However, the mechanism of its antidepressant effect has not been explored from the perspective of gut flora metabolism. Therefore, we employed both 16S rDNA gene sequencing and LC-MS metabolomics analysis to investigate the regulatory effects and mechanisms of lactoferrin in a rat model of depression. After one week of acclimatization, twenty-four 7-week-old male Sprague-Dawley rats were randomly and equally assigned into three groups: the control group, the model group, and the lactoferrin intervention group. The control group rats were housed under standard conditions, while the rats in the model and lactoferrin intervention groups were individually housed and exposed to chronic unpredictable mild stress for 44 days simultaneously. The lactoferrin intervention group was provided with water containing 2% lactoferrin (2 g/100 ml). Behavioural tests were conducted at week 7. Upon completion of the behavioral tests, the rats were anesthetized with isoflurane, humanely euthanized using a rat guillotine, and tissue samples were collected for further experiments. The results indicated that lactoferrin intervention led to an increase in sucrose solution consumption, horizontal movement distance, number of cross platforms, and residence time in the target quadrant. Additionally, it resulted in an increase in jejunal tight junction protein ZO-1 expression and a suppression of serum expression of inflammatory factors, Lipopolysaccharide and Diamine oxidase. In summary, lactoferrin can regulate the metabolic disorder of intestinal flora, reduce intestinal permeability, and further regulate the metabolic balance of hippocampal tissues through the microbiota-gut-brain axis. This process ultimately alleviates the depression-like behavior in rats.


Subject(s)
Depression , Lactoferrin , Metabolomics , Rats, Sprague-Dawley , Animals , Lactoferrin/metabolism , Lactoferrin/pharmacology , Male , Depression/metabolism , Depression/drug therapy , Rats , Metabolomics/methods , Chromatography, Liquid/methods , RNA, Ribosomal, 16S/genetics , Gastrointestinal Microbiome/drug effects , Disease Models, Animal , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , DNA, Ribosomal/genetics , Hippocampus/metabolism , Mass Spectrometry , Liquid Chromatography-Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL