[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (256)

Search Parameters:
Keywords = chronic mild stress

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 367 KiB  
Article
Exploring the Levels of Stress, Anxiety, Depression, Resilience, Hope, and Spiritual Well-Being Among Greek Dentistry and Nursing Students in Response to Academic Responsibilities Two Years After the COVID-19 Pandemic
by Polyxeni Μangoulia, Antonia Kanellopoulou, Georgia Manta, Georgios Chrysochoou, Evangelos Dimitriou, Theodora Kalogerakou and Maria Antoniadou
Healthcare 2025, 13(1), 54; https://doi.org/10.3390/healthcare13010054 - 31 Dec 2024
Viewed by 301
Abstract
Background: Dentistry and nursing students experience significant anxiety, negatively impacting their well-being and academic performance. Objectives: This study aims to assess the prevalence and relationships of stress, anxiety, depression, resilience, hope, and spiritual well-being among dentistry and nursing students, identify demographic influences and [...] Read more.
Background: Dentistry and nursing students experience significant anxiety, negatively impacting their well-being and academic performance. Objectives: This study aims to assess the prevalence and relationships of stress, anxiety, depression, resilience, hope, and spiritual well-being among dentistry and nursing students, identify demographic influences and propose strategies to enhance resilience and well-being. Methods: This study surveyed 271 students attending Greece’s departments of dentistry and nursing at the National and Kapodistrian University of Athens, using an electronic questionnaire aimed to assess stress, anxiety, and depression (depression, anxiety, stress scale—DASS-21); resilience (resilience assessment questionnaire—RAQ8, brief resilience scale—BRS); hope (adult hope scale—AHS); and spiritual well-being (functional assessment of chronic illness therapy–spiritual well-being scale—FACIT-Sp-12). The survey also collected demographic data to identify factors influencing these variables. Statistical analyses, including hierarchical multiple linear regression and t-tests, were performed to analyze the relationships between variables. Results: The sample included 145 dentistry and 126 nursing students, with 68.6% female and 80.1% undergraduate. Half of the students reported mild or higher levels of stress (48.7%), anxiety (51.3%), and depression (53.5%). The prevalence of depression was the highest in our sample, followed by anxiety and stress. Higher family wealth was associated with reduced stress levels, while female undergraduate students reported higher levels of anxiety than their male counterparts. Hope was a strong predictor of resilience, but stress and worry had a negative correlation. Conclusions: Promoting students’ well-being and academic success requires effective stress-reduction and resilience-building techniques to improve students’ performance and support future healthcare professionals’ personal sustainability and holistic growth. Full article
(This article belongs to the Special Issue Towards Holistic Healthcare: Advancing Nursing and Medical Education)
18 pages, 3262 KiB  
Article
Nelumbo nucifera Petals Ameliorate Depressive-like Symptom and Cognitive Deficit in Unpredictable Chronic Mild Stress Mouse Model
by Juthamart Maneenet, Yutthana Chotritthirong, Ashraf M. Omar, Rattanathorn Choonong, Supawadee Daodee, Orawan Monthakantirat, Charinya Khamphukdee, Supaporn Pitiporn, Suresh Awale, Kinzo Matsumoto and Yaowared Chulikhit
Nutrients 2025, 17(1), 94; https://doi.org/10.3390/nu17010094 - 29 Dec 2024
Viewed by 418
Abstract
Background Chronic stress exposure has been widely recognized as a significant contributor to numerous central nervous system (CNS) disorders, leading to debilitating behavioral changes such as anxiety, depression, and cognitive impairments. The prolonged activation of the hypothalamic–pituitary–adrenal (HPA) axis during chronic stress disrupts [...] Read more.
Background Chronic stress exposure has been widely recognized as a significant contributor to numerous central nervous system (CNS) disorders, leading to debilitating behavioral changes such as anxiety, depression, and cognitive impairments. The prolonged activation of the hypothalamic–pituitary–adrenal (HPA) axis during chronic stress disrupts the neuroendocrine balance and has detrimental effects on neuronal function and survival. Nelumbo nucifera (N. nucifera) Gaertn., commonly known as the lotus flower, is a traditional medicinal plant consumed for its purported benefits on mental and physical well-being. Despite its traditional use, limited scientific evidence supports these claims. Methods The present study explores the effects of N. nucifera, commonly known as the lotus flower, on cognitive performance and stress resilience in a mouse model subjected to unpredictable chronic mild stress (UCMS). Results Daily treatment significantly improved cognitive performance, alleviated depressive-like behaviors, and normalized hypothalamic–pituitary–adrenal (HPA) axis activity, as indicated by a 60.97% reduction in serum corticosterone. At the molecular level, N. nucifera petals also downregulated serum- and glucocorticoid-inducible kinase 1 (SGK1) mRNA expression while upregulating brain-derived neurotrophic factor (BDNF) mRNA expression and cyclic-adenosine monophosphate (cAMP) responsive element-binding protein (CREB) mRNA expression in the hippocampus and frontal cortex. These normalizations are critical, as chronic stress dysregulates HPA axis function, exacerbating behavioral changes. Furthermore, a phytochemical analysis resulted in the isolation of five major compounds, kaempferol (1), trifolin (2), kaempferol-3-neohesperidoside (3), icariside D2 (4), and β-sitosterol (5), each demonstrating significant monoamine oxidase (MAO) inhibitory activity. Conclusions These compelling findings suggest that N. nucifera petals not only alleviate stress-induced mood and cognitive deficits but also offer a promising avenue for modulating the HPA axis and promoting neuroprotection via essential neurotrophic factors and enzymatic pathways. We advocate for its potential as a complementary and alternative medicine for effective stress management. Future investigations should further explore its mechanisms of action and evaluate its clinical applicability in stress-related disorders. Full article
(This article belongs to the Section Nutrition and Public Health)
Show Figures

Figure 1

Figure 1
<p>Schematic experiment protocols. Animals (<span class="html-italic">n</span> = 60) were divided into non-stressed and UCMS groups at week 0 (<span class="html-italic">n</span> = 12 mice per group). The UCMS groups received various stressors for 6 weeks. The UCMS mice were divided into four groups, which were daily administered with vehicle (0.5% SCMC/day), imipramine (20 mg/kg/day), and <span class="html-italic">Nelumbo nucifera</span> (NN) (100 and 500 mg/kg/day) for 3 weeks. In the sixth week, behavioral tests (modified Y-maze test, novel object recognition test, tail suspension test, and forced swimming test) were performed. After the behavioral tests, the animals were sacrificed to collect their blood and brains for neurochemical assessment.</p>
Full article ">Figure 2
<p>Structure of isolated compounds <b>1</b>–<b>5</b>.</p>
Full article ">Figure 3
<p>The effect of <span class="html-italic">N. nucifera</span> (NN) petals on anhedonia as measured by the sucrose preference test. After initiating the UCMS procedure, the amount of 2% sucrose consumed by each animal group was measured as an indicator of anhedonia behavior. Each line shows the mean ± S.E.M. (12 animals per group). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. the non-stressed group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. the vehicle-treated UCMS group.</p>
Full article ">Figure 4
<p>The effect of <span class="html-italic">N. nucifera</span> (NN) petals on cognitive function in the modified Y-maze test (<b>A</b>,<b>B</b>) and novel object recognition test (<b>C</b>,<b>D</b>). The time spent exploring the novel arm in the modified Y-maze test and the percentage of discrimination index of each animal group were measured 6 weeks after starting the UCMS procedure. Each column shows the mean ± S.E.M. (12 animals per group). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. the non-stressed group. *** <span class="html-italic">p</span> &lt; 0.001 vs. the vehicle-treated UCMS group.</p>
Full article ">Figure 5
<p>The effect of <span class="html-italic">N. nucifera</span> (NN) petals on hopeless behaviors in the forced swimming test (<b>A</b>,<b>B</b>) and tail suspension test (<b>C</b>,<b>D</b>). Six weeks after UCMS, each animal group’s immobility times were measured as an index of learned helplessness. Each column shows the mean ± S.E.M. (12 animals per group). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. the non-stressed group. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. the vehicle-treated UCMS group.</p>
Full article ">Figure 6
<p>The effect of <span class="html-italic">N. nucifera</span> (NN) petals on serum corticosterone levels. Each column shows the mean ± S.E.M. (5 animals per group). <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. non-stressed group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. the vehicle-treated UCMS group.</p>
Full article ">Figure 7
<p>The effects of <span class="html-italic">N. nucifera</span> (NN) petals on the mRNA expression of SGK1 (<b>A</b>), CREB (<b>B</b>), and BDNF (<b>C</b>) in the frontal cortex and hippocampus. Each column shows the mean ± S.E.M. (5 animals per group). <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. the vehicle-treated non-stressed group. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. the vehicle-treated UCMS group. <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05 compared to <span class="html-italic">N. nucifera</span> petal doses.</p>
Full article ">
26 pages, 6218 KiB  
Article
Revealing the Mechanism of Hemerocallis citrina Baroni in Depression Treatment Through Integrated Network Pharmacology and Transcriptomic Analysis
by Shan Gao, Jihui Lu, Yixiao Gu, Yaozhi Zhang, Cheng Wang, Feng Gao, Ziqi Dai, Shujing Xu, Jindong Zhang, Yuqin Yang and Haimin Lei
Pharmaceuticals 2024, 17(12), 1704; https://doi.org/10.3390/ph17121704 - 17 Dec 2024
Viewed by 377
Abstract
Background/Objectives: Hemerocallis citrina Baroni (HCB) is a traditional herb for the treatment of depression in China. However, the active constituents and the underlying mechanisms of its antidepressant effects remain unclear. The aim of this study was to identify the bioactive constituents of [...] Read more.
Background/Objectives: Hemerocallis citrina Baroni (HCB) is a traditional herb for the treatment of depression in China. However, the active constituents and the underlying mechanisms of its antidepressant effects remain unclear. The aim of this study was to identify the bioactive constituents of HCB and elucidate its underlying mechanism for the treatment of depression. Methods: The constituents of HCB were systematically analyzed using UHPLC-Q-Orbitrap HRMS. Its antidepressant effect was evaluated by chronic unpredictable mild stress (CUMS)-induced depression. The mechanism of HCB in treating depression was investigated through network pharmacology and molecular docking. Subsequently, its potential mechanism for the treatment of depression was carried out by RNA sequencing. Finally, the mechanism was further verified by Western blot. Results: A total of 62 chemical constituents were identified from HCB using UHPLC-Q-Orbitrap HRMS, including 17 flavonoids, 11 anthraquinones, 11 alkaloids, 10 caffeoylquinic acid derivatives, five phenolic acids, five triterpenoids, and three phenylethanosides, 13 of which were identified as potential active constituents targeting 49 depression-associated proteins. Furthermore, HCB was found to significantly reduce cognitive impairment, anxiety-like behavior, and anhedonia-like behavior. The expression levels of 5-hydroxytryptamine (5-HT), dopamine (DA), and brain-derived neurotrophic factor (BDNF) were elevated in the hippocampal CA3 region. Results from network pharmacology and transcriptomics indicated that the PI3K/Akt/CREB signaling pathway is essential for the therapeutic effects of HCB on depression. Research in the field of molecular biology has conclusively demonstrated that HCB is associated with an increase in the expression levels of several important proteins. Specifically, there was a notable upregulation of phosphorylated PI3K (p-PI3K) relative to its unphosphorylated form PI3K, as well as an elevation in the ratio of phosphorylated Akt (p-Akt) to total Akt. Additionally, the study observed increased levels of phosphorylated CREB (p-CREB) compared to its unphosphorylated CREB. Conclusions: This study provides compelling evidence that HCB possesses the ability to mitigate the symptoms of depression through its influence on the PI3K/Akt/CREB signaling pathway. HCB could be developed as a promising therapeutic intervention for individuals struggling with depression, offering new avenues for treatment strategies that target this particular signaling mechanism. Full article
(This article belongs to the Special Issue Discovery of Novel Antidepressants and Anxiolytics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Mass spectrogram of HCB. (<b>A</b>) TICC of HCB obtained in ESI+ mode. Product ion spectra of (<b>B</b>) kaempferol-3-rutinoside. (<b>C</b>) quercetin. (<b>D</b>) rutin. (<b>E</b>) kwanzoquinone G. (<b>F</b>) rhein. (<b>G</b>) gallic acid. (<b>H</b>) clionasterol. (<b>I</b>) adenosine.</p>
Full article ">Figure 2
<p>HCB improved CUMS mice depression-like behaviors. (<b>A</b>) Schematic diagram of experimental design. (<b>B</b>) Representative images of movement trajectory. (<b>C</b>) Total distance within 5 min in the OFT (<span class="html-italic">n</span> = 8). (<b>D</b>) Time spent in the central area in the OFT (<span class="html-italic">n</span> = 8). (<b>E</b>) Not moving time in the OFT within 5 min (<span class="html-italic">n</span> = 8). (<b>F</b>) Immobility time in the FST within 4 min (<span class="html-italic">n</span> = 8). (<b>G</b>) Changes in precent of sucrose preference in the SPT (<span class="html-italic">n</span> = 8). (<b>H</b>) The secretion levels of 5-hydroxytryptamine (<span class="html-italic">n</span> = 3). (<b>I</b>) The secretion levels of dopamine (<span class="html-italic">n</span> = 3). (<b>J</b>) The secretion levels of BDNF (<span class="html-italic">n</span> = 3). (<b>K</b>) The number of Nissl bodies in the hippocampal CA3 regions (<span class="html-italic">n</span> = 3). (<b>L</b>) Representative pictures of Nissl staining in the hippocampi. Data are presented as mean ± SEM, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. control group (C-group); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. model group (M-group).</p>
Full article ">Figure 3
<p>Analysis results of network pharmacology and molecular docking. (<b>A</b>) Venn mapping of HCB on depression. (<b>B</b>) PPI networks of candidate targets. (<b>C</b>) The network construction of compounds–targets–diseases. (<b>D</b>) GO enrichment analysis. (<b>E</b>) KEGG pathway analysis. (<b>F</b>) Molecular docking diagram of active constitutes and potential targets.</p>
Full article ">Figure 4
<p>RNA sequencing analysis of hippocampus. (<b>A</b>) Volcano map of DEGs. (<b>B</b>) Hierarchical clustering analysis of DEGs. (<b>C</b>) Functional annotation analysis of GO using DEGs. (<b>D</b>) Functional enrichment analysis of KEGG using DEGs.</p>
Full article ">Figure 5
<p>HCB regulated PI3K/Akt/CREB signaling pathway (<b>A</b>) Representative protein bands of PI3K, p-PI3K, Akt, p-Akt, CREB, and p-CREB in hippocampal. Statistical graphs of relative protein expression of ratio of p-PI3K/PI3K (<b>B</b>), PI3K/GAPDH (<b>C</b>), p-Akt/Akt (<b>D</b>), Akt/GAPDH (<b>E</b>), p-CREB/CREB (<b>F</b>), and CREB/GAPDH (<b>G</b>). Data are presented as mean ± SEM, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. control group (C-group); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. model group (M-group).</p>
Full article ">Figure 6
<p>Identification of antidepressant constitutes in HCB and its underlying mechanism on the treatment of depression.</p>
Full article ">
12 pages, 267 KiB  
Review
The Role of Dietary Magnesium in Cardiovascular Disease
by Forrest H. Nielsen
Nutrients 2024, 16(23), 4223; https://doi.org/10.3390/nu16234223 - 6 Dec 2024
Viewed by 1949
Abstract
In the past 20 years, a large number of epidemiological studies, randomized controlled trials, and meta-analyses have found an inverse relationship between magnesium intake or serum magnesium and cardiovascular disease, indicating that low magnesium status is associated with hypertension, coronary artery calcification, stroke, [...] Read more.
In the past 20 years, a large number of epidemiological studies, randomized controlled trials, and meta-analyses have found an inverse relationship between magnesium intake or serum magnesium and cardiovascular disease, indicating that low magnesium status is associated with hypertension, coronary artery calcification, stroke, ischemic heart disease, atrial fibrillation, heart failure, and cardiac mortality. Controlled metabolic unit human depletion–repletion experiments found that a mild or moderate magnesium deficiency can cause physiological and metabolic changes that respond to magnesium supplementation, which indicates that these types of deficiencies or chronic latent magnesium deficiency are contributing factors to the occurrence and severity of cardiovascular disease. Mechanisms through which a mild or moderate magnesium deficiency can contribute to this risk include inflammatory stress, oxidative stress, dyslipidemia and deranged lipid metabolism, endothelial dysfunction, and dysregulation of cellular ion channels, transporters, and signaling. Based on USA official DRIs or on suggested modified DRIs based on body weight, a large number of individuals routinely consume less magnesium than the EAR. This especially occurs in populations that do not consume recommended amounts of whole grains, pulses, and green vegetables. Thus, inadequate magnesium status contributing to cardiovascular disease is widespread, making magnesium a nutrient of public health concern. Full article
(This article belongs to the Special Issue The Role of Magnesium Status in Human Health)
21 pages, 7804 KiB  
Article
Systemic Oxidative Stress Correlates with Sarcopenia and Pruritus Severity in Primary Biliary Cholangitis (PBC): Two Independent Relationships Simultaneously Impacting the Quality of Life—Is the Low Absorption of Cholestasis-Promoted Vitamin D a Puzzle Piece?
by Marcello Dallio, Mario Romeo, Fiammetta Di Nardo, Carmine Napolitano, Paolo Vaia, Lorenzo Ventriglia, Annachiara Coppola, Simone Olivieri, Marco Niosi and Alessandro Federico
Livers 2024, 4(4), 656-676; https://doi.org/10.3390/livers4040045 - 6 Dec 2024
Viewed by 570
Abstract
Background: Unlike other chronic liver disorders, in primary biliary cholangitis (PBC), systemic oxidative stress (SOS) worsens along with liver disease progression status (DPS), influencing muscle metabolism, muscle quantity (MQ), and itch pathways. Synergistically, cholestasis contributes to reduced vitamin D absorption, with a negative [...] Read more.
Background: Unlike other chronic liver disorders, in primary biliary cholangitis (PBC), systemic oxidative stress (SOS) worsens along with liver disease progression status (DPS), influencing muscle metabolism, muscle quantity (MQ), and itch pathways. Synergistically, cholestasis contributes to reduced vitamin D absorption, with a negative impact on MM and SOS. Despite this evidence, the prevalence of sarcopenia in PBC, and the SOS-MQ relationship comparing PBC with other CLDs, has never been investigated. Moreover, the relationship between vitamin D and MQ-SOS, and the correlation between SOS and pruritus severity, remains unexplored in PBC. Methods: A total of 40 MASLD, 52 chronic HBV infections, 50 chronic HCV infections, and 41 ursodeoxycholic acid/antioxidant-naïve PBC patients were enrolled. Biochemical, nutritional, and liver stiffness (LSM) data were collected, and sarcopenia was assessed after a normalizing 3-month dietetic–physical exercise regimen. The d-ROMs/BAP tests evaluated SOS. The validated “PBC-40 questionnaire” estimated pruritus and quality of life (QoL). Results: Unlike other CLDs, in PBC patients, sarcopenia was more prevalent in initial mild fibrosis (PBC: 57.10% vs. MASLD: 30.76%, HBV: 22.60%, HCV: 20.70%, all p < 0.0001), and SOS significantly correlated with MQ (dROMs-ASM/h2, p: 0.0002; BAP-ASM/h2: p: 0.0092). PBC patients presented lower vitamin D levels and a significant correlation of these with SOS and MQ (all p < 0.0001). SOS also correlated with pruritus severity (dROMs, R: 0.835; BAP, R: −0.775, p < 0.0001). QoL impairment was significantly more represented in PBC individuals with sarcopenia, SOS imbalance, and relevant pruritus (p: 0.0228). Conclusions: In PBC, SOS correlates with MQ impairment and pruritus severity, configuring two independent relationships simultaneously impacting QoL. Full article
Show Figures

Figure 1

Figure 1
<p>Experimental design. Anthropometrical parameters’ collection enclosed the determination of body mass index (BMI) (Kg/m<sup>2</sup>), weight (Kg), and height (cm). Clinical data included the complete medical history collection, drug abuse, comorbidities, pruritus quality of life (QoL) evaluation, and the concomitant therapies record. Nutritional counseling was offered to all the patients; dietary and physical exercise habits were assessed for this purpose. Body composition analysis and sarcopenia assessment were conducted via bioelectrical impedance analysis (BIA). Liver stiffness measurement (LSM) was the non–invasive tool (NIT) used to define liver disease progression status. Biochemical variables collected included aspartate aminotransferase (AST), alanine aminotransferase (ALT), platelet count (PLT), gamma–glutamyl transferase (GGT), alkaline phosphatase (ALP), albumin, total bilirubin (TB), International Normalized Ratio (INR), and vitamin D levels. TB ≥ 2 mg/dL distinguished severe cholestasis from moderate cholestasis. PBC: primary biliary cholangitis; CLDs: chronic liver disorders; HCV: Hepatitis C virus infection; HBV: Hepatitis B virus infection; MASLD: metabolic dysfunction–associated steatotic liver disease; SOS: systemic oxidative stress status; UDCA: ursodeoxycholic acid.</p>
Full article ">Figure 2
<p>The prevalence of sarcopenia according to liver DPS in PBC (<b>A</b>), MASLD (<b>B</b>), HBV (<b>C</b>), and HCV (<b>D</b>). The prevalence of sarcopenia in patients with initial mild/moderate fibrosis (F0–F2) across various CLD etiologies (<b>E</b>). DPS: disease progression status; MASLD: metabolic dysfunction–associated steatotic liver disease; HBV: chronic Hepatitis B infection; HCV: chronic Hepatitis C infection. Chi-square test. Statistically significant differences (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>The relationship between ASM/h<sup>2</sup> (MQ) and LSM (liver fibrosis severity) in PBC (<b>A</b>), MASLD (<b>B</b>), and viral-related CLD (HBV and HCV) (<b>C</b>). ASM: appendicular skeletal muscle mass; LSM: liver stiffness measurement; MASLD: metabolic dysfunction–associated steatotic liver disease; HBV: chronic Hepatitis B infection; HCV: chronic Hepatitis C infection. CLD: chronic liver disease. Linear regression analysis. Statistically significant differences (<span class="html-italic">p</span> &lt; 0.05) are in bold.</p>
Full article ">Figure 4
<p>The prevalence of SOS-I in PBC patients with severe and moderate cholestasis (<b>A</b>). The prevalence of sarcopenia in PBC patients with severe and moderate cholestasis (<b>B</b>). Chi-square test analysis. PBC: primary biliary cholangitis; SOS-I: systemic oxidative stress imbalance. Statistically significant differences (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Cholestasis, SOS, sarcopenia, and QoL in PBC patients. The pruritus severity (<b>A</b>) and the QoL impairment levels (<b>B</b>) in PBC patients overall and in PBC individuals with severe vs. moderate cholestasis. Mann–Whitney test analysis. The relationship of pruritus severity (<b>C</b>) and QoL impairment levels (<b>D</b>) with SOS (dROM and BAP) in PBC patients. Linear regression analysis. The prevalence of global QoL impairment in PBC individuals simultaneously presenting relevant pruritus, SOS–I, and sarcopenia compared with subjects presenting no relevant pruritus, no SOS-I, and no sarcopenia (<b>E</b>). Chi-square test analysis. PBC: primary biliary cholangitis; SOS: systemic oxidative stress; dROMs: derivatives of reactive oxidative metabolites; BAP: biological antioxidant potential; QoL: quality of life. Statistically significant differences (<span class="html-italic">p</span> &lt; 0.05) are in bold.</p>
Full article ">Figure 5 Cont.
<p>Cholestasis, SOS, sarcopenia, and QoL in PBC patients. The pruritus severity (<b>A</b>) and the QoL impairment levels (<b>B</b>) in PBC patients overall and in PBC individuals with severe vs. moderate cholestasis. Mann–Whitney test analysis. The relationship of pruritus severity (<b>C</b>) and QoL impairment levels (<b>D</b>) with SOS (dROM and BAP) in PBC patients. Linear regression analysis. The prevalence of global QoL impairment in PBC individuals simultaneously presenting relevant pruritus, SOS–I, and sarcopenia compared with subjects presenting no relevant pruritus, no SOS-I, and no sarcopenia (<b>E</b>). Chi-square test analysis. PBC: primary biliary cholangitis; SOS: systemic oxidative stress; dROMs: derivatives of reactive oxidative metabolites; BAP: biological antioxidant potential; QoL: quality of life. Statistically significant differences (<span class="html-italic">p</span> &lt; 0.05) are in bold.</p>
Full article ">Figure 6
<p>Vitamin D levels in the study population and correlation with SOS and MQ in PBC. The vitamin D levels in PBC patients vs. other CLDs (<b>A</b>) and in PBC patients with severe vs. moderate cholestasis (<b>B</b>). Mann–Whitney test analysis. The relationship between vitamin D levels and SOS (dROM and BAP) in PBC patients (<b>C</b>). The relationship between vitamin D levels and MQ (ASM/h<sup>2</sup>) in PBC patients (<b>D</b>). Linear regression analysis. PBC: primary biliary cholangitis; ASM: appendicular skeletal mass; h<sup>2</sup>: the square of the height; SOS: systemic oxidative stress; dROMs: derivatives of reactive oxidative metabolites; BAP: biological antioxidant potential. Statistically significant differences (<span class="html-italic">p</span> &lt; 0.05) are in bold.</p>
Full article ">
19 pages, 5583 KiB  
Article
(-)-Syringaresinol Exerts an Antidepressant-like Activity in Mice by Noncompetitive Inhibition of the Serotonin Transporter
by Yingyao Wu, Jianxin Cai, Hanhe Liu, Chan Li, Qingfa Tang and Yuan-Wei Zhang
Pharmaceuticals 2024, 17(12), 1637; https://doi.org/10.3390/ph17121637 - 5 Dec 2024
Viewed by 503
Abstract
Background: Albizia julibrissin Durazz. is one of the most popular herbs used for depression treatment, but the molecular basis for its mechanism of action has not been fully addressed. Previously, we isolated and identified two lignan glycoside derivatives that were shown to noncompetitively [...] Read more.
Background: Albizia julibrissin Durazz. is one of the most popular herbs used for depression treatment, but the molecular basis for its mechanism of action has not been fully addressed. Previously, we isolated and identified two lignan glycoside derivatives that were shown to noncompetitively inhibit serotonin transporter (SERT) activity but with a relatively low inhibitory potency compared with those of conventional antidepressants. Methods: We characterized the pharmacological profile of the parental compound of these previously isolated lignan glycosides, (-)-syringaresinol (SYR), in inhibiting SERT by using biochemical, pharmacological, and behavioral approaches. Results: SYR, as a potent inhibitor, decreases SERT Vmax but with little change in Km for its fluorescent substrate. SYR was shown to block the conformational conversion essential for substrate transport by stabilizing SERT in an outward-open and inward-closed conformation. In addition, our molecular docking and biochemical validation demonstrated that SYR binds to an allosteric site in SERT and noncompetitively inhibits SERT transport and binding activity. Furthermore, administration of SYR was indicated to exert an antidepressant-like activity and to effectively attenuate chronic unpredictable mild stress (CUMS)-induced abnormalities in behaviors and synaptic protein expression in depressive animal models. Conclusions: This study not only provides molecular insights into the mechanism of action of A. julibrissin in the treatment of depression, but also opens up the possibility of development of a novel class of allosteric site-targeted therapeutic agents with an underlying mechanism of action different from that of conventional antidepressants. Full article
(This article belongs to the Special Issue Neuropharmacology of Plant Extracts and Their Active Compounds)
Show Figures

Figure 1

Figure 1
<p>SYR noncompetitively inhibited SERT activity. (<b>A</b>) The structure of SYR. SYR holds a two-fold symmetrical structure with two 2,6-dimethoxyphenyl groups located at both sides. (<b>B</b>) SYR inhibition of both APP<sup>+</sup> uptake and ASP<sup>+</sup> binding by hSERT. The cells stably expressing hSERT were used for measuring APP<sup>+</sup> uptake or ASP<sup>+</sup> binding activity without or with SYR treatment at the indicated concentrations, as described in <a href="#sec4-pharmaceuticals-17-01637" class="html-sec">Section 4</a>. The graph shows APP<sup>+</sup> uptake or ASP<sup>+</sup> binding activity relative to that measured in the absence of SYR. IC<sub>50</sub> values for SYR inhibition of APP<sup>+</sup> uptake or ASP<sup>+</sup> binding were 0.25 ± 0.01 μM or 0.96 ± 0.02 μM, respectively. These IC<sub>50</sub> values represent the mean ± SEM from multiple experiments (<span class="html-italic">n</span> ≥ 3). (<b>C</b>) Noncompetitive inhibition of APP<sup>+</sup> uptake by SYR. APP<sup>+</sup> uptake was measured with or without the addition of 0.25 μM SYR. K<sub>m</sub> and V<sub>max</sub> values represent the mean ± SEM from multiple experiments (<span class="html-italic">n</span> ≥ 3). (<b>D</b>) Time course of APP<sup>+</sup> efflux under various treatments. The cells preloaded with APP<sup>+</sup> were incubated with KRH buffer in the absence or presence of 5-HT (10 μM), SYR (10 μM), 5-HT + fluoxetine (10 μM + 10 μM), or 5-HT + SYR (10 μM + 10 μM), at 22 °C for the indicated time periods. After 3× rapid washing, the APP<sup>+</sup> fluorescence retained in the cells was measured. The graph shows APP<sup>+</sup> efflux expressed as a percentage of total preloaded APP<sup>+</sup> fluorescence. The experiment was repeated two more times with similar results. CTRL, control; Fluox, fluoxetine.</p>
Full article ">Figure 2
<p>SYR effects on SERT cell surface expression. (<b>A</b>,<b>B</b>) Immunocytochemistry analysis for SERT expression on the cell surface. The cells stably expressing C-terminal Flag-tagged hSERT were treated with fluoxetine or SYR, and then incubated with anti-Flag monoclonal M2 antibody after permeabilization using 0.1% Triton X-100. The immunofluorescence images (<b>A</b>) were captured by confocal microscopy. Fluorescence intensity for SERT expression on the cell surface was quantified after subtraction of the intracellular fluorescence intensity from the total fluorescence (<b>B</b>). Error bars represent SEM from multiple experiments (<span class="html-italic">n</span> = 3). (<b>C</b>,<b>D</b>) Biotinylation analysis for SERT expression on the cell surface. The cells stably expressing C-terminal Flag-tagged SERT were treated with fluoxetine or SYR. SERT expressed on the cell surface was biotinylated with sulfo-NHS-SS-biotin, captured with streptavidin-agarose beads, and analyzed by immunoblot with anti-Flag monoclonal M2 antibody. Immunoreactive bands for SERT (<b>C</b>) were visualized by chemiluminescence, and quantification of SERT expression on the cell surface (<b>D</b>) was performed after normalization to total SERT expression in the cells. Error bars represent SEM from multiple experiments (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>SYR stabilized an outward-open and inward-closed conformation of SERT. (<b>A</b>) MTSET concentration-dependent inhibition of APP<sup>+</sup> uptake under various treatments. The inhibition of APP<sup>+</sup> uptake by MTSET over a range of concentrations (0–1 mM) was measured in the cells stably expressing Y107C/C109A with or without the addition of 10 μM fluoxetine (Fluox), 5-HT, or SYR. The graph shows APP<sup>+</sup> uptake relative to that in the absence of both MTSET and ligand. (<b>B</b>) Rate constants for MTSET reactivity with Y107C/C109A. Error bars represent SEM from multiple experiments (<span class="html-italic">n</span> ≥ 3). ** <span class="html-italic">p</span> &lt; 0.01 compared with the rate constant in the control (MTSET alone). (<b>C</b>) MTSEA concentration-dependent inhibition of ASP<sup>+</sup> binding under various treatments. Inhibition of ASP<sup>+</sup> binding by MTSEA at the indicated concentrations was measured in digitonin-permeabilized cells stably expressing S277C/X5C with or without the addition of 10 μM fluoxetine (Fluox), 5-HT, or SYR. (<b>D</b>) Rate constants for MTSEA reactivity with S277C/X5C. Error bars represent SEM from multiple experiments (<span class="html-italic">n</span> ≥ 3). ** <span class="html-italic">p</span> &lt; 0.01 compared with the rate constant in the control.</p>
Full article ">Figure 4
<p>Molecular docking of SYR to the allosteric site and its binding pose in SERT. (<b>A</b>) Overall view of the hSERT–SYR–imipramine complex model in cartoon representation. SYR (red) in the allosteric binding (S2) site is depicted as spheres. Purple spheres represent the imipramine molecule bound in the central binding (S1) site. (<b>B</b>) Close-up of SYR binding in the S2 site. Residues that were proposed to interact with SYR are annotated and shown as yellow sticks. Black numbers on α-helices represent the transmembrane α-helix numbers in hSERT. (<b>C</b>) Comparison of the binding poses for vilazodone and SYR within the S2 site. The main chain positions of the bundle domain and scaffold domain of hSERT in the hSERT–imipramine–vilazodone complex structure (PDB ID, 7lWD) are shown in gold and gray, and vilazodone and SYR are shown as cyan and red sticks, respectively.</p>
Full article ">Figure 5
<p>SYR inhibition of APP<sup>+</sup> transport (<b>A</b>) or ASP<sup>+</sup> binding (<b>B</b>) by hSERT and its S2 site mutants. APP<sup>+</sup> transport or ASP<sup>+</sup> binding by WT-hSERT or its allosteric S2 mutants was measured over the indicated range of SYR concentrations as described in <a href="#sec4-pharmaceuticals-17-01637" class="html-sec">Section 4</a>. Nonspecific APP<sup>+</sup> transport or ASP<sup>+</sup> binding was measured by adding 100 μM fluoxetine. The graph shows SYR inhibition of APP<sup>+</sup> transport or ASP<sup>+</sup> binding by hSERT and its S2 mutants relative to that measured without SYR addition. IC<sub>50</sub> values calculated from nonlinear regression analyses were shown as the mean ± SEM from at least three experiments in <a href="#pharmaceuticals-17-01637-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 6
<p>Animal behavioral tests. (<b>A</b>) Schematic illustration of animal behavioral test timeline. Mice were exposed to CUMS for 3 weeks, as described in <a href="#sec4-pharmaceuticals-17-01637" class="html-sec">Section 4</a>. Fluoxetine (10 mg/kg) or SYR (5, 10, or 20 mg/kg) was administered (i.p.) once per day from day 7 to day 21. For the CUMS group, saline was given instead of drugs. NSFT was performed after 16–18 h food deprivation, followed by FST and TST. (<b>B</b>) NSFT. The latency time to food in NSFT was counted. (<b>C</b>) Home cage food consumption. Food consumed by individual mice in the home cage (g) within a 20 min period was measured after NSFT. (<b>D</b>) FST. The immobility time during a 2 to 6 min period in FST was measured. (<b>E</b>) TST. The immobility time during 2 to 6 min was counted. <span class="html-italic">n</span> = 8–10 mice/group. Bars represent the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 compared with the control or CUMS group, respectively, by one-way ANOVA followed by post hoc tests. ns, not significant.</p>
Full article ">Figure 7
<p>SYR effects on the expression of c-Fos and synaptic proteins in mPFC. (<b>A</b>) Representative immunohistochemical images of c-Fos expression in mPFC under the indicated treatments. Immunohistochemistry of c-Fos was performed with mPFC slices as described in <a href="#sec4-pharmaceuticals-17-01637" class="html-sec">Section 4</a>. The slices were re-probed with anti-DAPI antibody. Immunofluorescence images for c-Fos (green) and DAPI (blue) were acquired by confocal microscopy. (<b>B</b>) Quantitative analysis for c-Fos expression in mPFC with various treatments. Protein expression is expressed as a percentage of c-Fos fluorescence measured in the control group. Bars represent the mean ± SEM (<span class="html-italic">n</span> = 4). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 compared with the control or CUMS group by one-way ANOVA followed by post hoc tests, respectively. (<b>C</b>) Representative immunoblots of the synaptic proteins BDNF, PSD-95, GluA1, and vGluT1 with various treatments. (<b>D</b>–<b>G</b>) Quantification of the expression of the synaptic proteins BDNF (<b>D</b>), PSD-95 (<b>E</b>), GluA1 (<b>F</b>), and vGluT1 (<b>G</b>). The expression level is expressed as a percentage of the integrated density obtained from the control group. Bars represent the mean ± SEM (<span class="html-italic">n</span> = 4). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 by one-way ANOVA followed by post hoc tests.</p>
Full article ">Figure 8
<p>SYR effects on the expression of c-Fos and synaptic proteins in the hippocampus. (<b>A</b>) Representative immunohistochemical images of c-Fos expression in the hippocampus with the indicated treatments. (<b>B</b>) Quantitative analysis for hippocampal c-Fos expression with various treatments. Protein expression level is expressed as a percentage of c-Fos fluorescence measured in the control group. Bars represent the mean ± SEM (<span class="html-italic">n</span> = 4). ** <span class="html-italic">p</span> &lt; 0.01 compared with the control or CUMS group by one-way ANOVA followed by post hoc tests. (<b>C</b>) Representative immunoblots of the synaptic proteins BDNF, PSD-95, GluA1, and vGluT1 with various treatments. (<b>D</b>–<b>G</b>) Quantification of the expression of synaptic proteins BDNF (<b>D</b>), PSD-95 (<b>E</b>), GluA1 (<b>F</b>), and vGluT1 (<b>G</b>). The expression level is expressed as a percentage of the integrated density obtained from the control group. Bars represent the mean ± SEM (<span class="html-italic">n</span> = 4). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 by one-way ANOVA followed by post hoc tests.</p>
Full article ">
29 pages, 21147 KiB  
Article
Gut-Microbiota-Derived Butyric Acid Overload Contributes to Ileal Mucosal Barrier Damage in Late Phase of Chronic Unpredictable Mild Stress Mice
by Chen Wang, Mei Qiu, Shuo Wang, Jinjin Luo, Ling Huang, Qi Deng, Zhijia Fang, Lijun Sun and Ravi Gooneratne
Int. J. Mol. Sci. 2024, 25(23), 12998; https://doi.org/10.3390/ijms252312998 - 3 Dec 2024
Viewed by 523
Abstract
Intestinal mucosal barrier damage is regarded as the critical factor through which chronic unpredictable mild stress (CUMS) leads to a variety of physical and mental health problems. However, the exact mechanism by which CUMS induces intestinal mucosal barrier damage is unclear. In this [...] Read more.
Intestinal mucosal barrier damage is regarded as the critical factor through which chronic unpredictable mild stress (CUMS) leads to a variety of physical and mental health problems. However, the exact mechanism by which CUMS induces intestinal mucosal barrier damage is unclear. In this study, 14, 28, and 42 d CUMS model mice were established. The indicators related to ileal mucosal barrier damage (IMBD), the composition of the ileal microbiota and its amino acid (AA) and short-chain fatty acid (SCFA) metabolic functions, and free amino acid (FAA) and SCFA levels in the ileal lumen were measured before and after each stress period. The correlations between them are analyzed to investigate how CUMS induces intestinal mucosal barrier damage in male C57BL/6 mice. With the progression of CUMS, butyric acid (BA) levels decreased (14 and 28 d) and then increased (42 d), and IMBD progressively increased. In the late CUMS stage (42 d), the degree of IMBD is most severe and positively correlated with significantly increased BA levels (p < 0.05) in the ileal lumen and negatively correlated with significantly decreased FAAs, such as aspartic, glutamic, alanine, and glycine levels (p < 0.05). In the ileal lumen, the abundance of BA-producing bacteria (Muribaculaceae, Ruminococcus, and Butyricicoccus) and the gene abundance of specific AA degradation and BA production pathways and their related enzymes are significantly increased (p < 0.05). In addition, there is a significant decrease (p < 0.05) in the abundance of core bacteria (Prevotella, Lactobacillus, Turicibacter, Blautia, and Barnesiella) that rely on these specific AAs for growth and/or are sensitive to BA. These changes, in turn, promote further colonization of BA-producing bacteria, exacerbating the over-accumulation of BA in the ileal lumen. These results were validated by ileal microbiota in vitro culture experiments. In summary, in the late CUMS stages, IMBD is related to an excessive accumulation of BA caused by dysbiosis of the ileal microbiota and its overactive AA degradation. Full article
(This article belongs to the Special Issue New Insights into Gut Microbiota and Immunity)
Show Figures

Figure 1

Figure 1
<p>Effect of chronic unpredictable mild stress (CUMS) on physiological parameters and depression-like behavior in mice. (<b>A</b>) Body weight; (<b>B</b>) body weight growth rate per 3 d; (<b>C</b>) body weight growth rate per 14 d; (<b>D</b>) food intake; (<b>E</b>) viscera coefficient; (<b>F</b>) sucrose preference test (SPT); (<b>G</b>) time in the central area in the open field test (OFT); (<b>H</b>) immobility time in the tail suspension test (TST); (<b>I</b>) immobility time in the forced-swim test (FST). Data are shown as mean ± SEM (<span class="html-italic">n</span> = 15). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CON. Control (CON) group, chronic unpredictable mild stress (CUMS) group.</p>
Full article ">Figure 2
<p>Effect of chronic unpredictable mild stress (CUMS) on ileal mucosal barrier damage (IMBD), DAO and LPS concentrations, and serum inflammatory marker levels in mice on 14, 28, and 42 d. (<b>A</b>) Histological changes in the ileal tissue (hematoxylin and eosin stain, magnification ×200); (<b>B</b>) ratio of villi height to crypt depth; (<b>C</b>) mean number of goblet cells in the visual field; (<b>D</b>) IMBD score; (<b>E</b>) Diamine Oxidase (DAO); (<b>F</b>) lipopolysaccharide (LPS); (<b>G</b>) TNF-α; (<b>H</b>) IL-1β; (<b>I</b>) IL-6; (<b>J</b>) IL-10. Data are shown as mean ± SEM (<span class="html-italic">n</span> = 5). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CON.</p>
Full article ">Figure 3
<p>Effect of chronic unpredictable mild stress (CUMS) on ileal occludin, ZO-1, Muc2, and Olfactomedin 4 (Olfm4+) protein expression in mice. (<b>A</b>–<b>D</b>) Immunohistochemistry of occludin, ZO-1, Muc2, and Olfm4+ proteins in the ileal tissue in different groups; (<b>E</b>) positive areas of occludin in the ileum; (<b>F</b>) positive areas of ZO-1 in the ileum; (<b>G</b>) positive areas of Muc2 in the ileum; (<b>H</b>) positive cells for Olfm4+ in the ileum. Data are shown as mean ± SEM (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CON.</p>
Full article ">Figure 4
<p>Effect of chronic unpredictable mild stress (CUMS) on the diversity and composition of ileal gut microbiota in mice. (<b>A</b>) Venn diagram based on OTU; (<b>B</b>) Shannon’s index; (<b>C</b>) Simpson’s index; (<b>D</b>) principal coordinate analysis (PCoA) of gut microbiota. Gut microbiota changes at the (<b>E</b>) phylum and (<b>H</b>) genus levels. (<b>F</b>) Ratio of Firmicutes and Bacteroidota between experimental groups. (<b>G</b>) Horizontal clustering heat map of the gut microbiota. (<b>I</b>) Classification of unit groups based on LDA histograms for taxa showed a significant difference between groups (<span class="html-italic">p</span> &lt; 0.05). Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 10). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CON.</p>
Full article ">Figure 5
<p>Effects of chronic unpredictable mild stress (CUMS) on the abundance of 16 characteristic marker bacteria in the ileal microbiota in mice. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 10). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CON.</p>
Full article ">Figure 6
<p>Effects of chronic unpredictable mild stress (CUMS) on the gene abundance of the ileal microbiota AAs, sugars, and SCFAs metabolic pathways and related enzymes in mice. (<b>A</b>) Analysis of the metabolic prediction of the Gut Metabolic Module (GMM) module of the gut microbiota, (<b>B</b>) changes in gene abundance of amino acid (AA) metabolism pathway, (<b>C</b>) changes in gene abundance of sugar metabolism pathway, (<b>D</b>) changes in gene abundance of SCFA metabolism, (<b>E</b>) dynamic variations in gene abundance of AA degradation and butyric acid (BA) production metabolism-related enzymes. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 10). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CON.</p>
Full article ">Figure 7
<p>Effects of chronic unpredictable mild stress (CUMS) on free amino acid (FAA) and short-chain fatty acid (SCFA) levels in the mice ileal contents. (<b>A</b>) Mice FAAs levels in the ileal contents, (<b>B</b>) mice SCFA (acetic, propionic, isobutyric, N-butyric, isovaleric, N-valeric) concentrations in the ileal contents. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 6). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CON.</p>
Full article ">Figure 8
<p>Correlation analysis of the ileal content levels of free amino acid (FAA) and butyric acid (BA); the abundance of the key ileal marker bacteria; and the gene abundance of amino acid (AA), sugar, and short-chain fatty acid (SCFA) metabolic pathways and associated enzymes in the ileal microbiota. And correlation analysis of the BA levels in the ileal contents and ileal mucosal barrier indices. (<b>A</b>) Correlation between the key ileal marker bacteria and gene abundance of ileal microbiota AA, sugar, and SCFA metabolism. (<b>B</b>) Correlation between the gene abundance of ileal microbiota AA, sugar, and SCFA metabolism and the AA-degradation- and BA-production-metabolism-related enzymes. (<b>C</b>) Relationship between the key ileal gut marker bacteria, BA, and gene abundance of AA degradation and BA production metabolism. (<b>D</b>) Relationship between the key ileal marker bacteria, BA, FAAs, and gene abundance of AA degradation and BA production metabolism. (<b>E</b>) Correlation analysis of the BA levels in the ileal contents and ileal mucosal barrier indices. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. Leucine (Leu), lysine (Lys), aspartic acid (Asp), glutamic acid (Glu), serine (Ser), glycine (Gly), threonine (Thr), alanine (Ala), tyrosine (Tyr), valine (Val), methionine (Met), isoleucine (Ile), diamine oxidase (DAO), lipopolysaccharide (LPS), Zonula occludens-1 (ZO-1), Mucoprotein 2 (Muc2), and Olfactomedin 4 (Olfm4+).</p>
Full article ">Figure 9
<p>Growth curves, pH, butyric acid (BA) levels, free amino acid (FAA) consumption rates, and the composition of 42 d CON and CUMS ileal flora in vitro culture. (<b>A</b>) Growth curves, (<b>B</b>) pH, (<b>C</b>) BA levels, (<b>D</b>) FAAs consumption rates. PCoA analysis of CON 42 d and CUMS 42 d ileal flora in vitro culture at (<b>E</b>) 12 and (<b>F</b>) 24 h. Ileal microbiota in vitro culture changes at the phylum (<b>G</b>) and genus (<b>H</b>) levels. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 6). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. CON.</p>
Full article ">Figure 10
<p>Flowchart of the chronic unpredicted mild stress (CUMS) procedure.</p>
Full article ">
21 pages, 6640 KiB  
Article
Combining Network Pharmacology and Transcriptomic Strategies to Explore the Pharmacological Mechanism of Total Ginsenoside Ginseng Root and Its Impact on Antidepressant Effects
by Weijia Chen, Pengli Guo, Lili Su, Xiangjuan Guo, Meiling Shi, Jianan Geng, Ying Zong, Yan Zhao, Rui Du and Zhongmei He
Int. J. Mol. Sci. 2024, 25(23), 12606; https://doi.org/10.3390/ijms252312606 - 24 Nov 2024
Viewed by 669
Abstract
Depression is one of the most common neurological diseases, which imposes a substantial social and economic burden on modern society. The purpose of this study was to explore the mechanism of total ginsenoside ginseng root (TGGR) in the treatment of depression through a [...] Read more.
Depression is one of the most common neurological diseases, which imposes a substantial social and economic burden on modern society. The purpose of this study was to explore the mechanism of total ginsenoside ginseng root (TGGR) in the treatment of depression through a comprehensive strategy combining network pharmacology, transcriptomics, and in vivo experimental validation. The Traditional Chinese Medicine Systematic Pharmacology (TCMSP) database and literature were used to collect the main components and targets of TGGR. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were applied to explore the underlying mechanisms. In addition, the chronic unpredictable mild stress (CUMS)-induced C57BL/6 mouse model was used to evaluate the antidepressant activity of TGGR. The results showed that TGGR improved depression-like behavior in mice and increased the decrease in serum 5-hydroxytryptamine (5-HT) and brain-derived neurotrophic factor (BDNF) levels caused by CUMS. Combined network pharmacology and transcriptomic analysis showed that the AMP-activated kinase (AMPK) signaling pathway mainly enriched the core target. Immunohistochemistry, Western blotting, and reverse transcription quantitative polymerase chain reaction (RT-qPCR) were used to confirm whether TGGR exerts antidepressant effects by regulating this pathway. The results showed that TGGR has a regulatory impact on related proteins in the AMPK pathway, and the regulatory effect of TGGR on proteins was inhibited after the administration of related pathway inhibitors. In summary, total ginsenosides may regulate the AMPK signaling pathway and activate the sirtuin 1 (SIRT1) peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α) pathway to have therapeutic effects on depression. Full article
(This article belongs to the Special Issue Pathophysiology and Pharmacology in Psychiatry)
Show Figures

Figure 1

Figure 1
<p>Potential targets of TGGR antidepressants and enrichment analyses. (<b>A</b>) Ginsenoside and depression target Venn plots. (<b>B</b>) Construction of the intersection target PPI network map of ginsenoside. (<b>C</b>,<b>D</b>) Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses.</p>
Full article ">Figure 2
<p>(<b>A</b>) Effects of TGGR on body weight of mice. (<b>B</b>) The effect of TGGR on sucrose preference in mice. (<b>C</b>,<b>D</b>) Effects of TGGR administration on resting time of FST and TST mice. (<b>E</b>) Heat map of MWM mouse activity. (<b>F</b>) The residence time of mice in the MWM target quadrant. (<b>G</b>) The levels of serum 5-HT and MDA, determined by ELISA. (<b>H</b>) Apoptosis of hippocampal neurons in each group (Nissl staining).The second picture for each group is a larger picture inside the red dashed box of the first picture. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 were significantly different from the control group, and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from model group.</p>
Full article ">Figure 3
<p>(<b>A</b>) DEGs crossover results in the control group, CUMS group, and TGGR group. (<b>B</b>,<b>C</b>) DEG volcanic distribution map. Blue dots indicate downregulated mRNA; red dots indicate upregulated mRNA. (<b>D</b>) Heat map analysis of low- and high-expression DEG. (<b>E</b>) GO enrichment analysis was performed on DEGs’ cellular components, biological processes, and molecular functions. The first 10 items are shown in the figure. (<b>F</b>) The top 10 pathways of DEGs after performing KEGG enrichment.</p>
Full article ">Figure 4
<p>The key targets and pathways of TGGR in the prevention and treatment of depression and the preliminary validation of this target and pathway of action, analyzed with a combination of network pharmacology and transcriptomics. (<b>A</b>) Cross-targets of TGGR and depression in network pharmacology and cross-targets of transcriptomic differential genes. (<b>B</b>) Cross-target GO enrichment analysis. (<b>C</b>) The top 10 KEGG enrichment pathways for cross-targets. (<b>D</b>,<b>E</b>) Expression of SIRT1 and PGC-1α in mouse hippocampus (immunohistochemical staining). (<b>F</b>) Western blot analysis of AMPK, SIRT1, and PGC-1α in the hippocampus of mice in each group. Protein expression was normalized to β-actin for quantitative analysis, and its value expressed as an average (<b>G</b>). Normalization of the data to β-actin. Values are expressed as average ± SD. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group, and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from model group.</p>
Full article ">Figure 5
<p>(<b>A</b>) Changes in body weight of mice. (<b>B</b>) Changes in sucrose preference of mice. (<b>C</b>,<b>D</b>) Effects of TGGR administration on resting time of TST and FST mice. (<b>E</b>) Heat maps of MWM mouse activity. (<b>F</b>) Residence time of mice in the MWM target quadrant. (<b>G</b>,<b>H</b>) ELISA-detected serum 5-HT and MDA levels. (<b>I</b>) Apoptosis of hippocampal neurons in each group (Nissl staining). ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 was significantly different from model group, △ <span class="html-italic">p</span> &lt; 0.05 was significantly different from TGGR pair, ns &gt; 0.05 was not significantly different.</p>
Full article ">Figure 6
<p>(<b>A</b>) Ultrastructure of hippocampal neurons and mitochondria of mice in each group. (<b>B</b>) ATP content in mouse hippocampus. (<b>C</b>) ROS levels in mouse brain tissue. (<b>D</b>) Expression of SIRT1 and PGC-1α in mouse hippocampus (immunohistochemical staining). (<b>E</b>) Western blot analysis of SIRT1 and PGC-1α in the hippocampus of mice in each group. (<b>F</b>) Protein expression normalized to β-actin for quantitative analysis, and its value expressed as an average. (<b>G</b>) SIRT1 and PGC-1α mRNA levels detected by RT-qPCR. Data are normalized to β-actin. Values are expressed as average ± SD. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 was substantially different from model group, and <sup>△</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>△△</sup> <span class="html-italic">p</span> &lt; 0.01 were substantially different from TGGR group.</p>
Full article ">
18 pages, 20350 KiB  
Article
Paeoniflorin Inhibits the Activation of Microglia and Alleviates Depressive Behavior by Regulating SIRT1-NF-kB-NLRP3/Pyroptosis Pathway
by Xue Wang, Lili Su, Silu Liu, Zhongmei He, Jianming Li, Ying Zong, Weijia Chen and Rui Du
Int. J. Mol. Sci. 2024, 25(23), 12543; https://doi.org/10.3390/ijms252312543 - 22 Nov 2024
Viewed by 539
Abstract
Inflammation assumes a vital role in the pathogenesis of depression and in antidepressant treatment. Paeoniflorin (PF), a monoterpene glycoside analog possessing anti-inflammatory attributes, exhibits therapeutic efficacy on depression-like behavior in mice. The objective of this study was to evaluate the antidepressant effects of [...] Read more.
Inflammation assumes a vital role in the pathogenesis of depression and in antidepressant treatment. Paeoniflorin (PF), a monoterpene glycoside analog possessing anti-inflammatory attributes, exhibits therapeutic efficacy on depression-like behavior in mice. The objective of this study was to evaluate the antidepressant effects of PF on depression elicited by the chronic unpredictable mild stress (CUMS) model and the precise neural sequence associated with the inflammatory process. In this study, we established an in vivo mouse model induced by CUMS and an in vitro BV2 cell model induced by LPS+ATP. The mechanism of PF for depression was assessed by the SIRT1 selective inhibitor EX-527. The findings demonstrated that PF significantly alleviated the damage of BV2 cells treated with LPS and ATP, inhibited the generation of ROS, up-regulated the expression of SIRT1 mRNA, and down-regulated the expression of nuclear NF-κB, p65, NLRP3, Caspase-1 and GSDMD-N in vitro. In vivo, PF mitigated the depressive-like behavior induced by CUMS, reduced the number of neurons, and decreased the secretion of pro-inflammatory factors IL-1β, IL-6, and TNF-α in the hippocampus. Immunohistochemical results indicated that PF attenuated CUMS-induced hyperactivation of microglia. Moreover, the expression level of SIRT1 in the hippocampus was augmented, while the protein levels of NF-κB, p65, NLRP3, Caspase-1, IL-1β and GSDMD-N were diminished after PF treatment. Additionally, the selective inhibition of SIRT1 attenuated the therapeutic effect of PF on depression. These results imply that PF possesses antidepressant properties that rely on SIRT1 signaling to regulate NLRP3 inflammasome inactivation. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>PF inhibited the activation of apoptosis, inflammation, pyroptosis and ROS of BV2 cells induced by LPS+ATP. (<b>A</b>) Cell viability of BV2. (<b>B</b>–<b>E</b>) The content of LDH, Caspase-1, IL-1β and IL-18. (<b>F</b>) Hoechst/PI double staining (×400). (<b>G</b>) ROS staining (×400). (<b>H</b>) Quantitative analysis of PI/Hoechst and DCFH-DA. Data are presented as mean ± SEM. ## <span class="html-italic">p</span> &lt; 0.01, compared to control group; ** <span class="html-italic">p</span> &lt; 0.01, compared to LPS+ATP group.</p>
Full article ">Figure 2
<p>Effects of PF on protein levels of SIRT1, NF-κB and NLRP3 inflammasome in LPS+ATP treated BV2 cells. The relative mRNA expression level of (<b>A</b>) SIRT1, (<b>B</b>) NF-κB, (<b>C</b>) NLRP3, (<b>D</b>) ASC, (<b>E</b>) Caspase-1, (<b>F</b>) IL-1β, (<b>G</b>) GSDMD-N. Data are presented as mean ± SEM. ## <span class="html-italic">p</span> &lt; 0.01, compared to control group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, compared to LPS+ATP group, <span>$</span> <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 compared to PF+EX-527 group.</p>
Full article ">Figure 3
<p>PF improved depressive-like behaviors in CUMS mice. (<b>A</b>) Experimental procedures. (<b>B</b>) SPT, (<b>C</b>) TST, (<b>D</b>) FST, (<b>E</b>,<b>G</b>) OFT and (<b>F</b>,<b>H</b>) EPM. Data are presented as mean ± SEM. ## <span class="html-italic">p</span> &lt; 0.01, compared to control group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, compared to CUMS group.</p>
Full article ">Figure 4
<p>PF alleviated neuroinflammation and neuronal damage in CUMS mice. (<b>A</b>) The levels of IL-6, IL-1β, TNF-α. (<b>B</b>) Quantitative analysis of Nissl bodies positive cells. (<b>C</b>) The positive signal intensity of IBA1. (<b>D</b>) Nissl staining image (×200). (<b>E</b>) Immunohistochemical image of IBA1 (×200). Data are presented as mean ± SEM. ## <span class="html-italic">p</span> &lt; 0.01, compared to control group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, compared to CUMS group.</p>
Full article ">Figure 5
<p>PF inhibited the activation of NF-κB by activating the expression of SIRT1. (<b>A</b>) Representative Western blots. (<b>B</b>) Quantification of SIRT1 and NF-κB P65 expression levels and fluorescence intensity. (<b>C</b>) The fluorescence intensity of SIRT1 (×400). (<b>D</b>) The fluorescence intensity of NF-κB P65 in CA3 area of hippocampus (×400). Data are presented as mean ± SEM. ## <span class="html-italic">p</span> &lt; 0.01, compared to control group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, compared to CUMS group.</p>
Full article ">Figure 6
<p>PF inhibited the activation of NLRP3 inflammasome and its mediated pyroptosis. (<b>A</b>) Representative Western blots. (<b>B</b>) The quantitative analysis of the expression level of NLRP3, ASC, Caspase-1, IL-1β and GSDMD-N. (<b>C</b>) The positive signal intensity of NLRP3 (×200). (<b>D</b>) IHC quantitative analysis of NLRP3. (<b>E</b>) The fluorescence intensity of GSDMD-N/DAPI in CA3 area of hippocampus (×400). Data are presented as mean ± SEM. ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001, compared to control group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared to CUMS group.</p>
Full article ">Figure 7
<p>EX-527 blocked the inhibitory effect of PF on the activation of NF-κB, NLRP3 inflammasome and pyroptosis in CUMS model mice. (<b>A</b>) Representative Western blots. (<b>B</b>) Quantification of SIRT1, NF-kB-P65, NLRP3, Caspase-1 and GSDMD-N expression levels and quantification of SIRT1 and GSDMD-N fluorescence intensity. (<b>C</b>) The fluorescence intensity of SIRT1/DAPI in CA3 area of hippocampus (×400). (<b>D</b>) The fluorescence intensity of GSDMD-N/DAPI in CA3 area of hippocampus (×400). Data are presented as mean ± SEM. ## <span class="html-italic">p</span> &lt; 0.01, compared to control group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, compared to CUMS group, <span>$</span> <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 compared to PF+EX-527 group.</p>
Full article ">
17 pages, 10646 KiB  
Article
Neuronal TCF7L2 in Lateral Habenula Is Involved in Stress-Induced Depression
by Xincheng Li, Xiaoyu Liu, Jiaxin Liu, Fei Zhou, Yunluo Li, Ye Zhao, Xueyong Yin, Yun Shi and Haishui Shi
Int. J. Mol. Sci. 2024, 25(22), 12404; https://doi.org/10.3390/ijms252212404 - 19 Nov 2024
Viewed by 678
Abstract
Depression is a complex psychiatric disorder that has substantial implications for public health. The lateral habenula (LHb), a vital brain structure involved in mood regulation, and the N-methyl-D-aspartate receptor (NMDAR) within this structure are known to be associated with depressive behaviors. Recent research [...] Read more.
Depression is a complex psychiatric disorder that has substantial implications for public health. The lateral habenula (LHb), a vital brain structure involved in mood regulation, and the N-methyl-D-aspartate receptor (NMDAR) within this structure are known to be associated with depressive behaviors. Recent research has identified transcription factor 7-like 2 (TCF7L2) as a crucial transcription factor in the Wnt signaling pathway, influencing diverse neuropsychiatric processes. In this study, we explore the role of TCF7L2 in the LHb and its effect on depressive-like behaviors in mice. By using behavioral tests, AAV-mediated gene knockdown or overexpression, and pharmacological interventions, we investigated the effects of alterations in TCF7L2 expression in the LHb. Our results indicate that TCF7L2 expression is reduced in neurons within the LHb of male ICR mice exposed to chronic mild stress (CMS), and neuron-specific knockdown of TCF7L2 in LHb neurons leads to notable antidepressant activity, as evidenced by reduced immobility time in the tail suspension test (TST) and forced swimming test (FST). Conversely, the overexpression of TCF7L2 in LHb neurons induces depressive behaviors. Furthermore, the administration of the NMDAR agonist NMDA reversed the antidepressant activity of TCF7L2 knockdown, and the NMDAR antagonist memantine alleviated the depressive behaviors induced by TCF7L2 overexpression, indicating the involvement of NMDAR. These findings offer novel insights into the molecular mechanisms of depression, highlighting the potential of TCF7L2 as both a biomarker and a therapeutic target for depression. Exploring the relationship between TCF7L2 signaling and LHb function may lead to innovative therapeutic approaches for alleviating depressive symptoms. Full article
Show Figures

Figure 1

Figure 1
<p>TCF7L2 in LHb neurons was downregulated in CMS mice. (<b>A</b>) Timeline of the CMS and behavioral tests, including the OFT, the TST, the EPM, the FST, and the SPT. (<b>B</b>) Time spent in the center in the OFT, mean of Naive: 30.98, mean of CMS: 18.46. (<b>C</b>) Total distance traveled during the OFT, mean of Naive: 2773, mean of CMS: 3258. (<b>D</b>) Latency to the first immobility in the TST, mean of Naive: 108.07, mean of CMS: 106.71. (<b>E</b>) Total immobility time in the TST, mean of Naive: 51.2, mean of CMS: 138.1. (<b>F</b>) Time spent in the open arms in the EPM, mean of Naive: 54.63, mean of CMS: 42.79. (<b>G</b>) Latency to the first floating in the FST, mean of Naive: 96.36, mean of CMS: 85.21. (<b>H</b>) Total floating time in the FST, mean of Naive: 93, mean of CMS: 139.9. (<b>I</b>) Sucrose preference (%) of SPT, mean of Naive: 75.69, mean of CMS: 61.18, (<b>J</b>,<b>K</b>) Immunofluorescence staining of TCF7L2 in LHb neurons, mean of Naive: 37.34, mean of CMS: 17.47, (red: TCF7L2, green: NeuN, blue: DAPI, scale bar = 50 μm; n = 4). Comparison between the Naive and CMS groups was conducted using T-tests or Mann–Whitney U-tests. Data are expressed as means ± SEM. n = 14 per group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, versus the Naive group.</p>
Full article ">Figure 2
<p>TCF7L2 knockdown in the LHb neurons caused antidepressant activity in mice. (<b>A</b>) Schematic of the experimental design of AAV-mediated TCF7L2 knockdown in the LHb neurons of mice. (<b>B</b>) Verification of TCF7L2 knockdown efficiency using fluorescence staining, mean of AAV-sh-scrambled: 93, mean of AAV-sh-TCF7L2: 139.9 (red: TCF7L2, green: GFP, blue: DAPI, scale bar = 50 μm). (<b>C</b>) Sucrose preference (%) of SPT, mean of AAV-sh-scrambled: 67.24, mean of AAV-sh-TCF7L2: 62.92. (<b>D</b>) Latency to eat in the NSF test, mean of AAV-sh-scrambled: 45.1, mean of AAV-sh-TCF7L2: 19.5. (<b>E</b>) Total intake of food in the NSF test, mean of AAV-sh-scrambled: 0.3667, mean of AAV-sh-TCF7L2: 0.3625. (<b>F</b>) Latency to the first immobility in the TST, mean of AAV-sh-scrambled: 107.2, mean of AAV-sh-TCF7L2: 116.5. (<b>G</b>) Total immobility time in the TST, mean of AAV-sh-scrambled: 66.86, mean of AAV-sh-TCF7L2: 37.53. (<b>H</b>) Latency to the first floating in the FST, mean of AAV-sh-scrambled: 59, mean of AAV-sh-TCF7L2: 104.8. (<b>I</b>) Total floating time in the FST, mean of AAV-sh-scrambled: 125.9, mean of AAV-sh-TCF7L2: 32.81. (<b>J</b>) Time spent in the center in the OFT, mean of AAV-sh-scrambled: 17.17, mean of AAV-sh-TCF7L2: 15.25. (<b>K</b>) Total distance traveled during the OFT, mean of AAV-sh-scrambled: 4039, mean of AAV-sh-TCF7L2: 3870. (<b>L</b>) Recognition index of NOR test, mean of AAV-sh-scrambled: 66.21, mean of AAV-sh-TCF7L2: 62.33. (<b>M</b>) Sniffing index in trial 1 of the three-chamber SIT, mean of AAV-sh-scrambled: 75.78, mean of AAV-sh-TCF7L2: 76.87. (<b>N</b>) Total sniffing time in trial 1 of the three-chamber SIT, mean of AAV-sh-scrambled: 85.82, mean of AAV-sh-TCF7L2: 77.41. (<b>O</b>) Preference index in trial 2 of the three-chamber SIT, mean of AAV-sh-scrambled: 35.31, mean of AAV-sh-TCF7L2: 35.61. (<b>P</b>) Total sniffing time in trial 2 of the three-chamber SIT, mean of AAV-sh-scrambled: 67.45, mean of AAV-sh-TCF7L2: 64.59. (<b>Q</b>) Analysis of the correlation between the total floating time in FST and density of TCF7L2<sup>+</sup> cells in LHb/mm<sup>2</sup>. Comparison between the AAV-sh-Scrambled and AAV-sh-TCF7L2 groups was conducted using the <span class="html-italic">T</span>-test or Mann–Whitney U-test. Data are expressed as means ± SEM. n = 10–23 per group. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, versus the AAV-sh-Scrambled group.</p>
Full article ">Figure 3
<p>TCF7L2 overexpression in the LHb neurons led to depressive-like behavior in mice. (<b>A</b>) Schematic of the experimental design of AAV-mediated TCF7L2 overexpression in the LHb neurons of mice. (<b>B</b>) Verification of AAV injection point using fluorescence staining, mean of AAV-EGFP: 67.45, mean of AAV-TCF7L2: 64.59 (red: TCF7L2, green: GFP, blue: DAPI, scale bar = 50 μm). (<b>C</b>) Sucrose preference (%) of SPT, mean of AAV-EGFP: 73.5, mean of AAV-TCF7L2: 74. (<b>D</b>) Latency to eat in the NSF test, mean of AAV-EGFP: 45.33, mean of AAV-TCF7L2: 124.27. (<b>E</b>) Total intake of food in the NSF test, mean of AAV-EGFP: 0.22, mean of AAV-TCF7L2: 0.16. (<b>F</b>) Latency to the first immobility in the TST, mean of AAV-EGFP: 97, mean of AAV-TCF7L2:88.06. (<b>G</b>) Total immobility time in the TST, mean of AAV-EGFP: 60.39, mean of AAV-TCF7L2: 98.89. (<b>H</b>) Latency to the first floating in the FST, mean of AAV-EGFP: 87.67, mean of AAV-TCF7L2: 64.11. (<b>I</b>) Total floating time in the FST, mean of AAV-EGFP: 50.06, mean of AAV-TCF7L2: 117.4. (<b>J</b>) Time spent in the center in the OFT, mean of AAV-EGFP: 17.61, mean of AAV-TCF7L2: 17.9. (<b>K</b>) Total distance traveled during the OFT, mean of AAV-EGFP: 2235, mean of AAV-TCF7L2: 2147. (<b>L</b>) Recognition index of NOR test, mean of AAV-EGFP: 59.34, mean of AAV-TCF7L2: 62.5. (<b>M</b>) Sniffing index in trial 1 of the three-chamber SIT, mean of AAV-EGFP: 79.49, mean of AAV-TCF7L2: 82.19. (<b>N</b>) Total sniffing time in trial 1 of the three-chamber SIT, mean of AAV-EGFP: 75.94, mean of AAV-TCF7L2: 78.22. (<b>O</b>) Preference index in trial 2 of the three-chamber SIT, mean of AAV-EGFP: 29.86, mean of AAV-TCF7L2: 25.55. (<b>P</b>) Total sniffing time in trial 2 of the three-chamber SIT, mean of AAV-EGFP: 71.94, mean of AAV-TCF7L2: 76.83. Comparison between the AAV-EGFP and AAV-TCF7L2 groups was conducted using <span class="html-italic">T</span>-test or Mann–Whitney U-test. (<b>Q</b>) Analysis of the correlation between the total floating time in FST and density of TCF7L2<sup>+</sup> cells in LHb/mm<sup>2</sup>. Data are expressed as means ± SEM. n = 18 per group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, versus the AAV-sh-Scrambled group.</p>
Full article ">Figure 4
<p>NMDAR was involved in TCF7L2-mediated depressive-like behavior. (<b>A</b>) Experimental timeline for NMDAR agonist-NMDA administration and behavioral tests. (<b>B</b>) Effects of NMDA administration on latency to the first immobility in the TST with LHb neurons special TCF7L2 knockdown, mean of AAV-sh-scrambled + Saline: 106, mean of AAV-sh-scrambled + NMDA: 111.9, mean of AAV-sh-TCF7L2 + Saline: 111.1, mean of AAV-sh-TCF7L2 + NMDA: 108.8. (<b>C</b>) Total immobility time in the TST, mean of AAV-sh-scrambled + Saline: 57.75, mean of AAV-sh-scrambled + NMDA: 52.45, mean of AAV-sh-TCF7L2 + Saline: 8.143, mean of AAV-sh-TCF7L2 + NMDA: 62.63. (<b>D</b>) Effects of NMDA administration on latency to the first floating in the FST, mean of AAV-sh-scrambled + Saline: 72.36, mean of AAV-sh-scrambled + NMDA: 95.45, mean of AAV-sh-TCF7L2 + Saline: 110.9, mean of AAV-sh-TCF7L2 + NMDA: 102.6. (<b>E</b>) Total floating time in the FST, mean of AAV-sh-scrambled + Saline: 106.1, mean of AAV-sh-scrambled + NMDA: 120.7, mean of AAV-sh-TCF7L2 + Saline: 45, mean of AAV-sh-TCF7L2 + NMDA: 134.7. (<b>F</b>) Experimental timeline for NMDAR antagonist-memantine administration and behavioral tests. (<b>G</b>) Effects of memantine administration on latency to the first immobility in the TST with LHb neurons special TCF7L2 overexpression, mean of AAV-EGFP + Saline: 85.25, mean of AAV-EGFP + Memantine: 88.25, mean of AAV-TCF7L2 + Saline: 50.75, mean of AAV-TCF7L2 + Memantine: 80.05. (<b>H</b>) Total immobility time in the TST, mean of AAV-EGFP + Saline: 79.75, mean of AAV-EGFP + Memantine: 75.38, mean of AAV-TCF7L2 + Saline: 131.5, mean of AAV-TCF7L2 + Memantine: 72.38. (<b>I</b>) Effects of memantine administration on latency to the first floating in the FST, mean of AAV-EGFP + Saline: 91.5, mean of AAV-EGFP + Memantine: 100.8, mean of AAV-TCF7L2 + Saline: 35, mean of AAV-TCF7L2 + Memantine: 82.5. (<b>J</b>) Total floating time in the FST, mean of AAV-EGFP + Saline: 32.25, mean of AAV-EGFP + Memantine: 44.63, mean of AAV-TCF7L2 + Saline: 157.5, mean of AAV-TCF7L2 + Memantine: 77.88. Comparisons between groups were conducted using one-way ANOVA. Data are expressed as means ± SEM. n = 7–11 per group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
21 pages, 312 KiB  
Review
Mandibular Advancement Devices in Obstructive Sleep Apnea and Its Effects on the Cardiovascular System: A Comprehensive Literature Review
by Agnieszka Polecka, Jakub Nawrocki, Maria Alejandra Pulido and Ewa Olszewska
J. Clin. Med. 2024, 13(22), 6757; https://doi.org/10.3390/jcm13226757 - 10 Nov 2024
Viewed by 2354
Abstract
Background: Obstructive sleep apnea syndrome (OSA) is a chronic inflammatory disease characterized by endothelial dysfunction and cardiovascular complications. Continuous positive airway pressure (CPAP) is the standard treatment, hence poor adherence has prompted interest in mandibular advancement devices (MAD) as an alternative. This comprehensive [...] Read more.
Background: Obstructive sleep apnea syndrome (OSA) is a chronic inflammatory disease characterized by endothelial dysfunction and cardiovascular complications. Continuous positive airway pressure (CPAP) is the standard treatment, hence poor adherence has prompted interest in mandibular advancement devices (MAD) as an alternative. This comprehensive review aimed to explore the effects of MAD therapy on oxidative stress, inflammation, endothelial function, and its impact on the cardiovascular risk in OSA patients. Results: MAD therapy significantly reduces the apnea-hypopnea index (AHI), improves serum nitric oxide (NOx) concentrations, reduces oxidative stress markers, and enhances endothelial function. Animal studies indicated that MAD reduces myocardial fibrosis and attenuates inflammatory markers. While both CPAP and MADs improve endothelial function and heart rate variability, CPAP is more effective in reducing OSA severity. Nevertheless, MAD has higher compliance, contributing to its positive impact on cardiovascular function. Moreover, CPAP and MADs have similar effectiveness in reducing cardiovascular risk. Conclusions: MAD therapy is an effective alternative to CPAP, particularly for patients with mild to moderate OSA as well as those intolerant to CPAP. It offers significant improvements in endothelial function and oxidative stress. Further studies are needed to assess MAD therapy in comprehensive OSA management. Full article
14 pages, 3544 KiB  
Article
Study on the Mechanism of Dictyophora duplicata Polysaccharide in Reducing Depression-like Behavior in Mice
by Chenxi Yang, Jiaqi Chen, Jie Tang, Lanzhou Li, Yongfeng Zhang, Yu Li, Changchun Ruan and Chunyue Wang
Nutrients 2024, 16(21), 3785; https://doi.org/10.3390/nu16213785 - 4 Nov 2024
Viewed by 1006
Abstract
Background/Objectives: Depression is a prevalent worldwide mental health disorder that inflicts significant harm to individuals and society. Dictyophora duplicata is an edible fungus that contains a variety of nutrients, including polysaccharides. This study aims to investigate the monosaccharide composition and molecular weight of [...] Read more.
Background/Objectives: Depression is a prevalent worldwide mental health disorder that inflicts significant harm to individuals and society. Dictyophora duplicata is an edible fungus that contains a variety of nutrients, including polysaccharides. This study aims to investigate the monosaccharide composition and molecular weight of the Dictyophora duplicata polysaccharide (DDP-B1), followed by an exploration of its antidepressant effects in chronic unpredictable mild stress (CUMS) mice. Methods: Dictyophora duplicata was purified using a DEAE-52 column and an S-400 column to obtain DDP-B1. The monosaccharide composition and molecular weight of DDP-B1 were investigated via high-performance gel permeation chromatograph. Six-week-old C57BL/6 male mice were utilized for the CUMS modeling to evaluate the antidepressant efficacy of DDP-B1. Fluoxetine served as the positive control group. The depressive-like behaviors and brain pathology of mice were evaluated. Immunofluorescence (IF) staining, metabolomics analysis, and western blot were employed to further investigate the underlying mechanisms. Results: DDP-B1 significantly alleviated the depression-like behavior of CUMS mice and increased the expression of SYN and PSD-95 in the mice’s brains, which was further validated by western blot. Metabolomics analysis indicated a reduction in serum glutamate in CUMS mice following DDP-B1 treatment. Moreover, DDP-B1 treatment led to an increase in levels of GABAAR, BDNF, p-TrkB and p-p70S6K. Conclusions: DDP-B1 regulated abnormalities in the glutamatergic system, subsequently activated the BDNF-TrkB-mTOR pathway and mitigated the pathological manifestations of CUMS mice. This study validated the potential of DDP-B1 as an antidepressant medication and established a theoretical foundation for the development of fungi with similar properties. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Figure 1

Figure 1
<p>Purification of DDP-B1 and analysis of monosaccharide composition. (<b>A</b>) DDP was isolated and purified using a DEAE-52 column, with elution performed using 0, 0.1, and 0.3 M NaCl solutions. The fraction DDP-B was obtained by eluting with the 0.1 M NaCl solution. (<b>B</b>) DDP-B1 was purified utilizing an S-400 column. (<b>C</b>) Monosaccharide composition of DDP-B1. DDP-B1 was determined to consist of the monosaccharide 1 mannose (15.278 min), 2 glucuronic acid (22.678 min) and 3 glucose (31.762 min). (<b>D</b>) Monosaccharide standard curve. According to the retention time, they are 1 mannose (15.360 min), 2 glucosamine hydrochloride (19.458 min), 3 rhamnose (21.530 min), 4 glucuronic acid (22.808 min), 5 galacturonic acid (25.862 min), 6 D-galactosamine hydrochloride (29.795 min), 7 glucose (31.995), 8 galactose (36.423 min), 9 xylose (38.890 min), 10 L-arabinose (40.267 min) and 11 fucose (47.302 min). Black line: response value; red line: quantitative baseline; dark blue line: retention time; light blue line: division line.</p>
Full article ">Figure 2
<p>DDP-B1 alleviated CUMS-induced depression-like behaviors. (<b>A</b>) A schematic representation of the mice experimental procedure. After 8 weeks of modeling, mice were randomly divided into four groups as follows: Control, CUMS, CUMS + Flx, and CUMS + DDP-B1. (<b>B</b>) SPT. (<b>C</b>) FST. (<b>D</b>) TST. (<b>E</b>) OFT. Data were expressed as mean ± S.E.M. (<span class="html-italic">n</span> = 9). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. Ctrl mice; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CUMS mice.</p>
Full article ">Figure 3
<p>DDP-B1 mitigated synaptic damage in CUMS mice subjected to CUMS. (<b>A</b>) SYN and PSD-95 were analyzed using IF staining at a magnification of (200×; scale bar: 50 μm) in the CA1 and CA3 regions of the mice hippocampus. Blue: 4′,6-Diamidino-2′-phenylindole (DAPI); green: SYN; red: PSD-95. (<b>B</b>) Quantitative analysis for SYN and PSD-95 in IF staining. (<b>C</b>) Representative images of western blot for SYN, PSD-95 and 5HT<sub>2C</sub>R proteins in the mice. Results of quantitative analysis for the expression levels of (<b>D</b>) SYN, (<b>E</b>) PSD-95 and (<b>F</b>) 5HT<sub>2C</sub>R. Data were expressed as mean ± S.E.M. (<span class="html-italic">n</span> = 3). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. Ctrl mice; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CUMS mice.</p>
Full article ">Figure 4
<p>Metabolomics analysis of mouse serum. (<b>A</b>) OPLS-DA score analysis. (<b>B</b>) Venn analysis. (<b>C</b>) Heatmap of significantly altered metabolites among Ctrl, CUMS and CUMS + DDP-B1 groups. (<b>D</b>) KEGG enrichment pathway diagram (<span class="html-italic">n</span> = 5).</p>
Full article ">Figure 5
<p>DDP-B1 modulates the BDNF-TrkB-mTOR signaling pathway. (<b>A</b>) Representative images of western blot. The administration of DDP-B1 significantly enhanced the expression levels of (<b>B</b>) GABA<sub>A</sub>R, (<b>C</b>) BDNF, (<b>D</b>) p-TrkB and (<b>E</b>) p-p70S6K in the brains of CUMS mice. Data were expressed as mean ± S.E.M. (<span class="html-italic">n</span> = 3). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. Ctrl mice; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CUMS mice.</p>
Full article ">
17 pages, 1366 KiB  
Article
Inhibition of Glycolysis Alleviates Chronic Unpredictable Mild Stress Induced Neuroinflammation and Depression-like Behavior
by Bing Liu, Ke Dong, Xiaobing Chen, Huafeng Dong, Yun Zhao, Xue Wang, Zhaowei Sun, Fang Xie and Lingjia Qian
Brain Sci. 2024, 14(11), 1098; https://doi.org/10.3390/brainsci14111098 - 30 Oct 2024
Viewed by 1020
Abstract
Background: Growing evidence suggests that glucose metabolism plays a crucial role in activated immune cells, significantly contributing to the occurrence and development of neuroinflammation and depression-like behaviors. Chronic stress has been reported to induce microglia activation and disturbances in glucose metabolism in the [...] Read more.
Background: Growing evidence suggests that glucose metabolism plays a crucial role in activated immune cells, significantly contributing to the occurrence and development of neuroinflammation and depression-like behaviors. Chronic stress has been reported to induce microglia activation and disturbances in glucose metabolism in the hippocampus. Aims: This study aims to investigate how chronic stress-mediated glycolysis promotes neuroinflammation and to assess the therapeutic potential of the glycolysis inhibitor, 2-deoxy-D-glucose (2-DG), in a model of chronic stress-induced neuroinflammation and depression-like behavior. Methods: In in vitro studies, we first explored the effects of 2-DG on the inflammatory response of microglia cells. The results showed that corticosterone (Cort) induced reactive oxygen species (ROS) production, increased glycolysis, and promoted the release of inflammatory mediators. However, these effects were reversed by intervention with 2-DG. Subsequently, we examined changes in depression-like behavior and hippocampal glycolysis in mice during chronic stress. The results indicated that chronic stress led to prolonged escape latency in the Morris water maze, increased platform-crossing frequency, reduced sucrose preference index, and extended immobility time in the forced swim test, all of which are indicative of depression-like behavior in mice. Additionally, we found that the expression of the key glycolytic enzyme hexokinase 2 (HK2) was upregulated in the hippocampus of stressed mice, along with an increased release of inflammatory factors. Further in vivo experiments investigated the effects of 2-DG on glycolysis and pro-inflammatory mediator production, as well as the therapeutic effects of 2-DG on chronic stress-induced depression-like behavior in mice. The results showed that 2-DG alleviated chronic stress-induced depression-like behaviors, such as improving escape latency and platform-crossing frequency in the Morris water maze, and increasing the time spent in the center of the open field. Additionally, 2-DG intervention reduced the level of glycolysis in the hippocampus and decreased the release of pro-inflammatory mediators. Conclusions: These findings suggest that 2-DG can mitigate neuroinflammation and depressive behaviors by inhibiting glycolysis and inflammatory responses. Overall, our results highlight the potential of 2-DG as a therapeutic agent for alleviating chronic stress-induced neuroinflammation through the regulation of glycolysis. Full article
(This article belongs to the Section Neuropharmacology and Neuropathology)
Show Figures

Figure 1

Figure 1
<p>Chronic stress induces depressive-like behavior in mice and leads to the activation of neuroinflammation. (<b>A</b>) Schematic timeline of CUMS and behavior test. (<b>B</b>) Concentrations (ng/mg) of corticosterone in the hippocampus of control and stressed mice at the end of the CUMS procedure (n = 10, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>C</b>) Representative track images of mice in the probe trial of MWM. (<b>D</b>,<b>E</b>) Escaping latency and crossing-platform times of mice (n = 10, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>F</b>) Representative track images of mice in the Open Field Test (OFT) and time spent in the central (n = 10, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>G</b>–<b>I</b>) Chronic unpredictable mild stress (CUMS)-induced depression-like behaviors were assessed by sucrose preference in the sucrose preference test (SPT) (<b>G</b>), immobility time in the forced swimming test (FST) (<b>H</b>), and immobility time in the Tail Suspension Test (TST) (<b>I</b>) (n = 10, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>J</b>) Representative images of IF staining of hippocampal sections from control and CUMS mice. Iba-1, green; DAPI, blue. Scale bar, 50 μm. (<b>K</b>–<b>M</b>) Levels of IL-6, IL-1β, and TNF-α in hippocampus lysates from control and CUMS mice as determined by ELISA (n = 10, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 2
<p>Chronic stress induces the upregulation of key enzymes of glycolysis in the mouse hippocampus. (<b>A</b>) Glucose levels in hippocampus lysates from control and CUMS mice (n = 10, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>B</b>) HK activity in hippocampus lysates from control and CUMS mice (n = 6, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>C</b>) Western blot of the expression of HK2 and PKM2 protein in hippocampus lysates from control and CUMS mice (n = 3, Student’s <span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01). (<b>D</b>) qRT-PCR assays monitoring the expression of HK2 and PKM2 in hippocampal lysates from control and CUMS mice (n = 6, Student’s <span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>Exposure to high concentrations of corticosterone promotes the inflammatory response of microglia. (<b>A</b>) BV2 cells were treated with the indicated concentrations of Cort for 24 h, and cell growth was evaluated using a cell counting kit-8 (CCK-8) assay in three independent experiments; one-way ANOVA with Tukey’s post hoc test, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. (<b>B</b>) qRT-PCR assays monitoring the expression of inflammatory factors, IL-1β, IL-6, and TNF-α in BV2 cells incubated with Cort (50 µmol/L) for 24 h. Control cells were incubated with DMSO (n = 3, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>C</b>) Representative images of IL-β staining in Cort-treated BV2 cells and control cells. Scale bar, 50 μm. (<b>D</b>) BV2 cells were treated with Cort for 24 h. Intracellular ROS levels were quantified using DCFH-DA (n = 3, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>E</b>) qRT-PCR assays monitor the expression of inflammatory factors, IL-1β, IL-6, and TNF-α in primary microglia cells incubated with Cort (50 µmol/L) for 24 h. Control cells were incubated with DMSO (n = 3, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>Exposure to high corticosterone promotes increased glycolysis in microglia. (<b>A</b>–<b>J</b>) Western blot of the expression of HK2 protein in BV2 and primary microglia cells lysates from control and Cort. Control cells were incubated with DMSO (n = 3, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>B</b>,<b>K</b>) qRT-PCR assays monitoring the expression of HK2 level in BV2 and primary microglia cells incubated with Cort (50 µmol/L) for 24 h. (n = 3, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>C</b>,<b>L</b>) HK activity in BV2 and primary microglia cells lysates from control and Cort (n = 3, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>D</b>) Seahorse Extracellular Flux analysis to quantify ATP production in BV2 cells (n = 3, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>E</b>–<b>I</b>) Treatment with Cort for 24 h significantly inhibited OCR in BV2 cells. Treatment with Cort for 24 h significantly increased glycolytic capacity of BV2 cells. Seahorse Extracellular Flux analysis to quantify basal and maximal respiration in BV2 cells. All the experiments were performed by the Agilent’s Seahorse Bioscience XF24 Extracellular Flux Analyzer. (n = 3, Student’s <span class="html-italic">t</span>-test, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 5
<p>2-DG ameliorates corticosterone-induced microglia inflammation by inhibiting glycolysis. (<b>A</b>) BV2 cells were treated with the indicated concentrations of 2-DG for 24 h, and cell growth was evaluated using a cell counting kit-8 (CCK-8) assay in three independent experiments; one-way ANOVA with Tukey’s post hoc test, ns: <span class="html-italic">p</span> &gt; 0.05. (<b>B</b>) qRT-PCR assays monitoring the expression of HK2 level in BV2 cells incubated with Cort (50 µmol/L) or 2-DG (1 mmol/L) for 24 h. (n = 3, one-way ANOVA with Tukey’s post hoc test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>C</b>) HK activity in BV2 cells lysates from control, Cort, and Cort +2-DG (n = 3, one-way ANOVA with Tukey’s post hoc test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>D</b>–<b>F</b>) qRT-PCR assays monitoring the expression of inflammatory factors, IL-6, IL-1β, and TNF-α in BV2 cells incubated with Cort (50 µmol/L) or 2-DG (1 mmol/L) for 24 h. Control cells were incubated with DMSO (n = 3, one-way ANOVA with Tukey’s post hoc test, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>2-DG ameliorates chronic stress-induced neuroinflammation and depressive-like behavior in mice by inhibiting glycolysis. (<b>A</b>) Schematic timeline of CUMS, 2-DG treatment, and behavior test. (<b>B</b>) qRT-PCR assays monitoring the expression of HK2 in hippocampal lysates from control and CUMS mice (n = 6, one-way ANOVA with Tukey’s post hoc test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>C</b>–<b>E</b>) Levels of IL-1β, IL-6 and TNF-α in hippocampus lysates from control, CUMS, and CUMS+2-DG mice as determined by ELISA (n = 10, one-way ANOVA with Tukey’s post hoc test, ** <span class="html-italic">p</span> &lt; 0.01). (<b>F</b>–<b>H</b>) Representative track images of mice in the probe trial of MWM, escaping latency and crossing-platform time of mice (n = 10, one-way ANOVA with Tukey’s post hoc test, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
27 pages, 12640 KiB  
Article
20 (S)-Protopanaxadiol Alleviates DRP1-Mediated Mitochondrial Dysfunction in a Depressive Model In Vitro and In Vivo via the SIRT1/PGC-1α Signaling Pathway
by Pengli Guo, Zixian Wang, Li Sun, Zhongmei He, Jianming Li, Jianan Geng, Ying Zong, Weijia Chen and Rui Du
Molecules 2024, 29(21), 5085; https://doi.org/10.3390/molecules29215085 - 28 Oct 2024
Viewed by 870
Abstract
Depression is a complex and common mental illness affecting physical and psychological health. Panax ginseng C. A. Mey is a traditional Chinese medicine with abundant pharmacological activity and applications in regulating mood disorders. 20 (S)-Protopanaxadiol is the major intestinal metabolite of ginsenoside and [...] Read more.
Depression is a complex and common mental illness affecting physical and psychological health. Panax ginseng C. A. Mey is a traditional Chinese medicine with abundant pharmacological activity and applications in regulating mood disorders. 20 (S)-Protopanaxadiol is the major intestinal metabolite of ginsenoside and one of the active components in ginseng. In this study, we aimed to investigate the therapeutic effects of 20 (S)-Protopanaxadiol on neuronal damage and depression, which may involve mitochondrial dynamics. However, the mechanism underlying the antidepressant effects of 20 (S)-Protopanaxadiol is unelucidated. In the present study, we investigated the potential mechanisms underlying the antidepressant activity of 20 (S)-Protopanaxadiol by employing a corticosterone-induced HT22 cellular model and a chronic unpredicted mild stress (CUMS)-induced animal model in combination with a network pharmacology approach. In vitro, the results showed that 20 (S)-Protopanaxadiol ameliorated the corticosterone (CORT)-induced decrease in HT22 cell viability, decrease in 5-hydroxytryptamine (5-HT) levels, and increase in nitric oxide (NO) and malondialdehyde (MDA) levels. Furthermore, 20 (S)-Protopanaxadiol exerted improvement effects on the CORT-induced increase in HT22 cell mitochondrial reactive oxygen species, loss of mitochondrial membrane potential, and apoptosis. In vivo, the results showed that 20 (S)-Protopanaxadiol ameliorated depressive symptoms and hippocampal neuronal damage in CUMS mice, and sirtuin1 (SIRT1) and peroxisome proliferator-activated receptor-1-Alpha (PGC-1α) activity were activated in the hippocampus of mice, thereby alleviating mitochondrial dysfunction and promoting the clearance of damaged mitochondria. In both in vivo and in vitro models, after inhibiting SIRT1 expression, the protective effect of 20 (S)-Protopanaxadiol on mitochondria was significantly weakened, and dynamin-related protein 1 (DRP1)-mediated mitochondrial division was significantly reduced. These findings suggest that 20 (S)-Protopanaxadiol may exert neuroprotective and antidepressant effects by attenuating DRP1-mediated mitochondrial dysfunction and apoptosis by modulating the SIRT1/PGC-1α signaling pathway. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

Figure 1
<p>Enrichment analysis and molecular docking analysis of intersection genes between 20 (S)-Protopanaxadiol and depression. (<b>A</b>) Venn plots of Ginsenoside and depression intersection genes. (<b>B</b>) PPI network diagram of intersection genes. (<b>C</b>,<b>D</b>) Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. (<b>E</b>) Genes-pathway network. Purple graphics represent genes and green graphics represent pathways. (<b>F</b>) Molecular docking of 20 (S)-Protopanaxadiol to key targets (ERS1, PTGS2, and SIRT1).</p>
Full article ">Figure 2
<p>Effects of 20 (S)-Protopanaxadiol on cell viability, depression-related indicators, and mitochondrial function of HT22 cells. (<b>A</b>,<b>B</b>) Screening the optimal concentration range of 20 (S)-Protopanaxadiol and the optimal modeling concentration of CORT. (<b>C</b>) Effect of 20 (S)-Protopanaxadiol on CORT-induced HT22 cell viability. (<b>D</b>–<b>F</b>) Effects of 20 (S)-Protopanaxadiol on 5-HT, NO, and MDA contents in HT22 cells induced by CORT. (<b>G</b>) Effect of 20 (S)-Protopanaxadiol on mitochondrial ATP production in HT22 cells induced by CORT. (<b>H</b>) Effect of 20 (S)-Protopanaxadiol on mitochondrial content in HT22 cells induced by CORT. (<b>I</b>) Effect of 20 (S)-Protopanaxadiol on CORT-induced mitochondrial membrane potential in HT22 cells. (<b>J</b>) Effect of 20 (S)-Protopanaxadiol on ROS content in HT22 cells induced by CORT. All data are expressed as mean ± standard deviation. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 were significantly different from the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from the CORT group; □□ <span class="html-italic">p</span> &lt; 0.01 was significantly different from the 12.5 μM group; ◊ <span class="html-italic">p</span> &lt; 0.05 and ◊◊ <span class="html-italic">p</span> &lt; 0.01 were significantly different from the 25 μM group.</p>
Full article ">Figure 3
<p>Effects of 20 (S)-Protopanaxadiol on CORT-induced apoptosis and mitochondrial dynamics-related protein expression in HT22 cells. (<b>A</b>) Hoechst 33342 /PI staining. (<b>B</b>) Expression of SIRT1, PGC-1α, and DRP1 in HT22 cells. (<b>C</b>–<b>E</b>) Results of quantitative detection of protein expression. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from the CORT group.</p>
Full article ">Figure 4
<p>SIRT1 inhibitor EX-527 was used to examine the effects of 20 (S)-Protopanaxadiol on cell viability, depression-related indicators, mitochondrial content, and mitochondrial function. (<b>A</b>) Effect of 20 (S)-Protopanaxadiol on HT22 cell viability induced by CORT. (<b>B</b>) Effects of 20 (S)-Protopanaxadiol on 5-HT, NO, and MDA levels in HT22 cells induced by CORT. (<b>C</b>) Effect of 20 (S)-Protopanaxadiol on mitochondrial content of HT22 cells induced by CORT. (<b>D</b>) Effect of 20 (S)-Protopanaxadiol on mitochondrial membrane potential induced by CORT in HT22 cells. (<b>E</b>) Effect of 20 (S)-Protopanaxadiol on reactive oxygen species content in HT22 cells induced by CORT. (<b>F</b>–<b>H</b>) Quantitative results of mitochondrial content, mitochondrial membrane potential, and reactive oxygen species content, respectively. (<b>I</b>) Effect of 20 (S)-Protopanaxadiol on ATP content in mitochondria of HT 22 cells induced by cortisol. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 significantly different from CORT group; <sup>△</sup> <span class="html-italic">p</span> &lt;0.05 and <sup>△△</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from those of 20 (S)-Protopanaxadiol (50 μM) group.</p>
Full article ">Figure 5
<p>SIRT1 inhibitor EX-527 was used to examine the effects of 20 (S)-Protopanaxadiol on apoptosis and mitochondrial dynamics-related proteins. (<b>A</b>) Hoechst 33342 /PI staining. (<b>B</b>) Expression of SIRT1, PGC-1α, and DRP1 in HT22 cells. (<b>C</b>–<b>E</b>) Quantitative detection results of protein expression. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from control group; <sup>#</sup> <span class="html-italic">p</span> &lt;0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 significantly different from CORT group; <sup>△</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>△△</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from those of 20 (S)-Protopanaxadiol (50 μM) group.</p>
Full article ">Figure 6
<p>Effects of 20 (S)-Protopanaxadiol (labeled as 20 (S)-pro in the figure) on depression-like behavior. (<b>A</b>) Changes in body weight of mice in each group over 7 weeks. (<b>B</b>) Changes in sucrose preference rate of mice in each group within 7 weeks. (<b>C</b>) MWM mouse activity thermogram. (<b>D</b>) Dwell time of mice in MWM target quadrant. (<b>E</b>) Immobility time of mice in OFT. (<b>F</b>) Activity heat map of 8-arm maze mice. All data are expressed as mean ± standard deviation, <span class="html-italic">n</span> = 8. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from CUMS group.</p>
Full article ">Figure 7
<p>Effects of 20 (S)-Protopanaxadiol (labeled as 20 (S)-pro in the figure) on serum depression-related factors, hippocampus neurons, hippocampus mitochondria morphology, and mitochondria function in mice. (<b>A</b>,<b>B</b>) Determination of serum 5-HT, MDA content. (<b>C</b>) Determination of ATP content in the hippocampus. (<b>D</b>) Nissl staining results image of the hippocampus. (<b>E</b>) Electron microscope image of hippocampus tissue. All data are expressed as mean ± standard deviation. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from the model group.</p>
Full article ">Figure 8
<p>Effect of 20 (S)-Protopanaxadiol (labeled as 20 (S)-pro in the figure) on mitochondrial dynamics-related protein expression. (<b>A</b>) Immunohistochemical signal intensity of SIRT1 and PGC-1α in hippocampus. (<b>B</b>) Expression of SIRT1, PGC-1α, DRP1, and BDNF in hippocampus. (<b>C</b>–<b>F</b>) Results of quantitative detection of protein expression in sea. (<b>G</b>) Immunofluorescence images show the colocalization of DRP1 (green) and TOMM20 (red). The nuclei were stained with 2-(4-amidinophenyl)-6-indolecarbamidine dihydrochloride (DAPI). All data are expressed as mean ± standard deviation, <span class="html-italic">n</span> = 8. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from the model group.</p>
Full article ">Figure 9
<p>To verify the effects of 20 (S)-Protopanaxadiol (labeled as 20 (S)-pro in the figure) on mitochondrial ultrastructure and function. (<b>A</b>) Electron microscope image of hippocampus tissue. (<b>B</b>) Determination of ATP content in the hippocampus. (<b>C</b>) Determination of serum 5-HT content. (<b>D</b>) Immunohistochemical signal intensity of SIRT1 and PGC-1α in hippocampus. All data are expressed as mean ± standard deviation. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 were significantly different from the control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 were significantly different from the model group; and <sup>△</sup> <span class="html-italic">p</span> &lt; 0.05 was significantly different from the 20 (S)-pro group.</p>
Full article ">Figure 10
<p>To verify the effect of 20 (S)-Protopanaxadiol (labeled as 20 (S)-pro in the figure) on mitochondrial dynamics-related protein expression. (<b>A</b>) Expression of SIRT1, PGC-1α, and DRP1 in hippocampus. (<b>B</b>–<b>D</b>) Results of quantitative detection of protein expression in sea. (<b>E</b>) Immunofluorescence images show the colocalization of DRP1 (green) and TOMM20 (red). All data are expressed as mean ± standard deviation. ** <span class="html-italic">p</span> &lt; 0.01 was significantly different from the control group; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 was significantly different from the model group; <sup>△△</sup> <span class="html-italic">p</span> &lt; 0.01 was significantly different from the 20 (S)-pro group.</p>
Full article ">
14 pages, 244 KiB  
Article
Demographics, Clinical Characteristics, and Well-Being of Veterans with TBI and Dementia and Their Caregivers
by Linda O. Nichols, Jennifer Martindale-Adams, Ronald T. Seel, Jeffrey K. Zuber and Paul B. Perrin
Geriatrics 2024, 9(5), 130; https://doi.org/10.3390/geriatrics9050130 - 8 Oct 2024
Cited by 1 | Viewed by 1237
Abstract
Background: This study provides a detailed examination of older Veterans with traumatic brain injury (TBI) and dementia and their caregivers, focusing on Veterans’ demographic, clinical, functional, safety risk, and behavioral characteristics and caregivers’ demographic, clinical, and care-related characteristics and well-being. Methods: Veterans’ caregivers [...] Read more.
Background: This study provides a detailed examination of older Veterans with traumatic brain injury (TBI) and dementia and their caregivers, focusing on Veterans’ demographic, clinical, functional, safety risk, and behavioral characteristics and caregivers’ demographic, clinical, and care-related characteristics and well-being. Methods: Veterans’ caregivers (N = 110) completed a telephone-based survey. Results: Veterans averaged eight comorbid health conditions, with over 60% having chronic pain, hypertension, post-traumatic stress disorder, or depression. Caregivers reported helping with an average of three activities of daily living, with the highest percentages of Veterans needing assistance with grooming, dressing, and bathing. Almost all Veterans needed assistance with shopping, cooking, medication management, housework, laundry, driving, and finances. Veterans averaged two safety risks, the most common being access to dangerous objects, access to a gun, and not being able to respond to emergency situations. Although Veterans averaged 14 behavioral concerns, caregivers reported that their family needs relating to TBI were generally met or partly met, and they voiced confidence in their ability to respond to behaviors and control their upsetting thoughts. Caregivers’ mean burden score was severe, while mean depression and anxiety scores were mild. Caregivers reported an average of 10.5 h per day providing care and 20.1 h per day on duty. Conclusions: The findings demonstrate the increased presence of impairments, safety risks, and behavioral issues in Veterans with comorbid TBI and dementia, as well as increased impacts on families’ burdens and care provision requirements. Clinicians should be alert for and educate TBI patients and caregivers on the warning signs of post-TBI dementia and its associated functional, behavioral, and safety risk profile, as well as challenges related to caregiver well-being. Healthcare policymakers must consider the increased caregiver demands associated with comorbid TBI and dementia, as well as the need for expanded long-term support and services. Full article
Back to TopTop