[go: up one dir, main page]

 
 
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

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
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (652)

Search Parameters:
Keywords = depression-like behavior

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 4185 KiB  
Article
Sex-Specific Behavioral and Molecular Responses to Maternal Lipopolysaccharide-Induced Immune Activation in a Murine Model: Implications for Neurodevelopmental Disorders
by Jing Xu, Rujuan Zhao, Mingyang Yan, Meng Zhou, Huanhuan Liu, Xueying Wang, Chang Lu, Qiang Li, Yan Mo, Paihao Zhang, Xingda Ju and Xianlu Zeng
Int. J. Mol. Sci. 2024, 25(18), 9885; https://doi.org/10.3390/ijms25189885 - 13 Sep 2024
Viewed by 261
Abstract
Maternal immune activation (MIA) during pregnancy has been increasingly recognized as a critical factor in the development of neurodevelopmental disorders, with potential sex-specific impacts that are not yet fully understood. In this study, we utilized a murine model to explore the behavioral and [...] Read more.
Maternal immune activation (MIA) during pregnancy has been increasingly recognized as a critical factor in the development of neurodevelopmental disorders, with potential sex-specific impacts that are not yet fully understood. In this study, we utilized a murine model to explore the behavioral and molecular consequences of MIA induced by lipopolysaccharide (LPS) administration on embryonic day 12.5. Our findings indicate that male offspring exposed to LPS exhibited significant increases in anxiety-like and depression-like behaviors, while female offspring did not show comparable changes. Molecular analyses revealed alterations in pro-inflammatory cytokine levels and synaptic gene expression in male offspring, suggesting that these molecular disruptions may underlie the observed behavioral differences. These results emphasize the importance of considering sex as a biological variable in studies of neurodevelopmental disorders and highlight the need for further molecular investigations to understand the mechanisms driving these sex-specific outcomes. Our study contributes to the growing evidence that prenatal immune challenges play a pivotal role in the etiology of neurodevelopmental disorders and underscores the potential for sex-specific preventative approaches of MIA. Full article
(This article belongs to the Special Issue Molecular Investigations in Neurodevelopmental Disorders)
Show Figures

Figure 1

Figure 1
<p>LPS-induced MIA male offspring showed elevated anxiety in elevated plus maze test. (<b>A</b>) Schematic of the experimental design. Pregnant C57BL/6J mice were i.p. injected with lipopolysaccharide (LPS) (60 μg/kg) or Phosphate Buffered Saline (PBS) on E12.5 to induce maternal immune activation (MIA). E12.5, Embryonic day 12.5. Red box: the center of the chamber. (<b>B</b>) Maternal serum concentrations of IL-6 (PBS, <span class="html-italic">n</span> = 5, LPS, <span class="html-italic">n</span> = 4) at 3 h after PBS or LPS injection into pregnant dams at E12.5. Unpaired Student’s <span class="html-italic">t</span>-tests. *** <span class="html-italic">p</span> &lt; 0.001, Mean ± SEM. red line: the mean value. (<b>C</b>) Relative IL-6 mRNA expression in E12.5 at 3 h after PBS and LPS injection in placentas of pregnant mice (PBS, <span class="html-italic">n</span> = 10, LPS, <span class="html-italic">n</span> = 13). Unpaired Student’s <span class="html-italic">t</span> test. ** <span class="html-italic">p</span> &lt; 0.01. Mean ± SEM. (<b>D</b>) Heatmap of mouse movement in the elevated plus maze test in 5 min. (<b>E</b>) Number of entries to the close zone. (<b>F</b>) Time in the close zone. (<b>G</b>) Number of entries to the open zone. (<b>H</b>) Time in the open zone. (<b>E</b>–<b>H</b>) <span class="html-italic">n</span> = 16 per group; two-way ANOVA with Tukey’s post hoc tests. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. Means ± SEM.</p>
Full article ">Figure 2
<p>LPS-induced MIA male offspring showed elevated anxiety in open field test. (<b>A</b>) Heatmap of mouse movement in the open-field test in 5 min. (<b>B</b>) Average speed. (<b>C</b>) Total distance traveled. (<b>D</b>) Time in the center zone. (<b>E</b>) Total distance traveled in the center zone. (<b>F</b>) Time in the outside zone. (<b>G</b>) Total distance traveled in the outside zone. <span class="html-italic">n</span> = 18 per group; two-way ANOVA with Tukey’s post hoc tests. * <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; 0.0001. Means ± SEM. LPS, lipopolysaccharide. MIA, maternal immune activation.</p>
Full article ">Figure 3
<p>LPS-induced MIA male offspring showed elevated levels of depression in tail suspension test. (<b>A</b>) Total time mobile. (<b>B</b>) Maximum movement speed. <span class="html-italic">n</span> = 17 per group; two-way ANOVA with Tukey’s post hoc tests. ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001. Means ± SEM. LPS, lipopolysaccharide. MIA, maternal immune activation.</p>
Full article ">Figure 4
<p>Maternal weight and offspring development in response to LPS exposure. (<b>A</b>) Maternal weight changes across embryonic days 12.5 to 18.5 post-PBS or LPS treatment. <span class="html-italic">n</span> = 4–19 per group, two-way ANOVA with Tukey’s post hoc tests. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. Mean ± SEM. (<b>B</b>) Litter size differences between PBS and LPS groups. <span class="html-italic">n</span> = 5–7 per group, Mann–Whitney U test. (<b>C</b>) Offspring weight progression by sex. <span class="html-italic">n</span> = 4–15 per group, two-way ANOVA with Tukey’s post hoc tests. PBS-Female vs. LPS-Female, * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. PBS-Male vs. LPS-Male, ++++ <span class="html-italic">p</span> &lt; 0.0001. Means ± SEM. LPS, lipopolysaccharide. MIA, maternal immune activation.</p>
Full article ">Figure 5
<p>LPS-induced MIA did not lead to noticeable alterations in the size of the offspring’s brain. (<b>A</b>) Schematic representation of brain statistics of PBS and LPS offspring. The horizontal blue arrow represents the width of brain, the vertical blue arrow indicates the length of brain, the cortical area is marked with a red line, and the red arrow signifies the length of the cortex. Scale bar = 5 mm. (<b>B</b>) Brain of PBS and LPS adult offspring. (<b>C</b>) Length of brain. (<b>D</b>) Width of brain. E Length of cortex. PBS male, <span class="html-italic">n</span> = 20; PBS female, <span class="html-italic">n</span> = 20; LPS male, <span class="html-italic">n</span> = 14; LPS female, <span class="html-italic">n</span> = 14; two-way ANOVA with Tukey’s post hoc tests. Means ± SEM. LPS, lipopolysaccharide. MIA, maternal immune activation.</p>
Full article ">Figure 6
<p>LPS-induced MIA promoted significant alterations in the synaptic-related genes of male offspring. (<b>A</b>) Luminex to measure the protein expression levels of various cytokines in the cortex of P0 offspring mice in PBS-M group, LPS-M group, PBS-F group, and LPS-F group (<span class="html-italic">n</span> = 3–5 per group). (<b>B</b>) PCR array experiments showing differential expression of genes associated with synaptic plasticity in the cortex of P0 offspring of mice of PBS-M, LPS-M, PBS-F, and LPS-F group. (<b>C</b>) Selected Gene ontology (GO) annotations enriched in the LPS-M and PBS-M groups. (<b>D</b>) Selected Gene ontology (GO) annotations enriched in the LPS-F and PBS-F groups. Bar plot shows the top 10 enrichment score [−log<sub>10</sub> (Q-value)] of DEGs involving a biological process. (<b>E</b>) Venn diagram showing shared and unique upregulated genes in the KEGG pathways within the cortices of PBS and LPS offspring of different sexes. (<b>F</b>) Venn diagram showing shared and unique downregulated genes in the KEGG pathways within the cortices of PBS and LPS offspring of different sexes. PBS-M, PBS-Male; LPS-M, LPS-Male; PBS-F, PBS-Female and LPS-F, LPS-Female. LPS, lipopolysaccharide. MIA, maternal immune activation.</p>
Full article ">
14 pages, 905 KiB  
Article
Intakes of Lean Proteins and Processed Meats and Differences in Mental Health between Rural and Metro Adults 50 Years and Older
by Nathaniel R. Johnson, Sherri N. Stastny and Julie Garden-Robinson
Nutrients 2024, 16(18), 3056; https://doi.org/10.3390/nu16183056 - 11 Sep 2024
Viewed by 495
Abstract
Mental health disparities exist between rural and metro areas of the United States. Differences in dietary intake may contribute to these disparities. We examined differences in dietary intake and mental health between those 50 years and older (n = 637) living in [...] Read more.
Mental health disparities exist between rural and metro areas of the United States. Differences in dietary intake may contribute to these disparities. We examined differences in dietary intake and mental health between those 50 years and older (n = 637) living in rural counties to those living in metro counties in North Dakota and the relationship between dietary intake to days with depression or anxiety. A survey was conducted throughout North Dakota. Items were modified from other surveys, such as the Behavioral Risk Factor Surveillance System questionnaires and the National Health Interview Survey Cancer Control Supplement Dietary Screener Questionnaire. Comparing medians, individuals more likely to be unable to perform normal daily activities due to mental health (p = 0.009) resided in rural areas instead of metro areas. Those living rurally also ate more processed meats (p = 0.005), while trending toward less lean protein intake (p = 0.056). Multinomial regression analyses controlling for covariates revealed that lean protein intake and fruit intake were inversely associated with days with depression and anxiety (all p < 0.05), whereas processed meat intake was positively associated with anxiety (p = 0.005). Clinicians working with older adults residing in rural areas should emphasize substituting lean proteins for processed meats. Full article
(This article belongs to the Special Issue Animal-Originated Food Intake and Human Health)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The interaction between self-reported days with depression and days with anxiety, with the number of self-reported “not good” mental health days. A mixed general linear model was used (Model: R<sub>2</sub> = 0.501, F<sub>29,559</sub> = 19.365, <span class="html-italic">p</span> &lt; 0.001; interaction: F<sub>13,559</sub> = 1.970, <span class="html-italic">p</span> = 0.021; main effect of depression: F<sub>5,559</sub> = 9.720, <span class="html-italic">p</span> &lt; 0.001; main effect of anxiety = 5.606 <span class="html-italic">p</span> &lt; 0.001). Data are estimated marginal means on a 1 to 6 ordinal scale (1 = 0–5 days, 2 = 6–10 days, 3 = 11–15 days, 4 = 16–20 days, 5 = 21–25 days, and 6 = 26–30 days) controlling for age, sex, race and ethnicity, cohabitation status, education level, and household income as covariates.</p>
Full article ">Figure 2
<p>The interaction between self-reported days with depression and days with anxiety, with the number of self-reported days being unable to do normal activities due to mental health. A mixed general linear model was used (Model: R<sup>2</sup> = 0.328, F<sub>29,559</sub> = 9.417, <span class="html-italic">p</span> &lt; 0.001; interaction: F<sub>13,559</sub> = 1.849, <span class="html-italic">p</span> = 0.033; main effect of depression: F<sub>5,559</sub> = 7.214, <span class="html-italic">p</span> &lt; 0.001; main effect of anxiety = 2.304, <span class="html-italic">p</span> = 0.043). Data are estimated marginal means on a 1 to 6 ordinal scale (1 = 0–5 days, 2 = 6–10 days, 3 = 11–15 days, 4 = 16–20 days, 5 = 21–25 days, and 6 = 26–30 days) controlling for age, sex, race and ethnicity, cohabitation status, education level, and household income as covariates.</p>
Full article ">
14 pages, 338 KiB  
Article
Grade-Level Differences in the Profiles of Substance Use and Behavioral Health Problems: A Multi-Group Latent Class Analysis
by Kechna Cadet, Ashley V. Hill, Tamika D. Gilreath and Renee M. Johnson
Int. J. Environ. Res. Public Health 2024, 21(9), 1196; https://doi.org/10.3390/ijerph21091196 - 10 Sep 2024
Viewed by 285
Abstract
We investigated associations between polysubstance use and behavioral problems among adolescents. Because substance use becomes more developmentally normative with age, we examined whether polysubstance use was less likely to co-occur with behavioral problems among older (vs. younger) adolescents. Using data from a nationally [...] Read more.
We investigated associations between polysubstance use and behavioral problems among adolescents. Because substance use becomes more developmentally normative with age, we examined whether polysubstance use was less likely to co-occur with behavioral problems among older (vs. younger) adolescents. Using data from a nationally representative survey of US high school students, we compared the association between polysubstance use (i.e., use of alcohol, cannabis, tobacco/nicotine, and illicit drugs) and behavioral problems (i.e., suicide attempts, depressive symptoms, poor school performance, and sexual risk behaviors) by grade level. We conducted latent class analysis (LCA) to characterize patterns of polysubstance use, and multi-group LCA to estimate invariance by grade. Among the three latent classes that emerged, classes were distinguished by having low, moderate, and high probabilities for behavior problems and use of substances. Class I comprised 52% of the sample, whereas classes II and III comprised 35% and 12% of the sample, respectively. The multi-group LCA showed that younger adolescents had a higher relative probability of co-occurring problem behaviors and polysubstance use. Findings may be helpful in targeting screening and prevention efforts of high school students by grade. Specifically, our results provide evidence that associations between behavioral problems and alcohol/drug use are weaker in later high school grades, suggesting that substance use may not be a weaker marker of behavioral problems for students in higher grades. Full article
(This article belongs to the Special Issue Alcohol and Drugs of Addiction, Aggression and Violence)
31 pages, 2179 KiB  
Review
Advancing Post-Stroke Depression Research: Insights from Murine Models and Behavioral Analyses
by Mădălina Iuliana Mușat, Bogdan Cătălin, Michael Hadjiargyrou, Aurel Popa-Wagner and Andrei Greșiță
Life 2024, 14(9), 1110; https://doi.org/10.3390/life14091110 - 3 Sep 2024
Viewed by 340
Abstract
Post-stroke depression (PSD) represents a significant neuropsychiatric complication that affects between 39% and 52% of stroke survivors, leading to impaired recovery, decreased quality of life, and increased mortality. This comprehensive review synthesizes our current knowledge of PSD, encompassing its epidemiology, risk factors, underlying [...] Read more.
Post-stroke depression (PSD) represents a significant neuropsychiatric complication that affects between 39% and 52% of stroke survivors, leading to impaired recovery, decreased quality of life, and increased mortality. This comprehensive review synthesizes our current knowledge of PSD, encompassing its epidemiology, risk factors, underlying neurochemical mechanisms, and the existing tools for preclinical investigation, including animal models and behavioral analyses. Despite the high prevalence and severe impact of PSD, challenges persist in accurately modeling its complex symptomatology in preclinical settings, underscoring the need for robust and valid animal models to better understand and treat PSD. This review also highlights the multidimensional nature of PSD, where both biological and psychosocial factors interplay to influence its onset and course. Further, we examine the efficacy and limitations of the current animal models in mimicking the human PSD condition, along with behavioral tests used to evaluate depressive-like behaviors in rodents. This review also sets a new precedent by integrating the latest findings across multidisciplinary studies, thereby offering a unique and comprehensive perspective of existing knowledge. Finally, the development of more sophisticated models that closely replicate the clinical features of PSD is crucial in order to advance translational research and facilitate the discovery of future effective therapies. Full article
(This article belongs to the Special Issue Feature Paper in Physiology and Pathology)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the literature search using PubMed.</p>
Full article ">Figure 2
<p>Diagram showing risk factors associated with PSD. Serotonin transporter (SERT), brain-derived neurotrophic factor (BNF), apolipoprotein E (ApoE), methylenetetrahydrofolate reductase (MTHFR), and Protein Kinase C Eta Gene (PRKCH).</p>
Full article ">Figure 3
<p>Diagram showing murine models of PSD. Bilateral Common Carotid Artery Occlusion (BCCAO), chronic unpredictable mild stress (CUMS), Middle Cerebral Artery Occlusion (MCAO), 5-Hydroxytryptamine (5-HT, serotonin), and brain-derived neurotrophic factor (BDNF).</p>
Full article ">Figure 4
<p>Behavioral assessment methods for post-stroke depression in rodent models.</p>
Full article ">
18 pages, 1173 KiB  
Review
Obstructive Sleep Apnea and Serotoninergic Signalling Pathway: Pathomechanism and Therapeutic Potential
by Alicja Witkowska, Julia Jaromirska, Agata Gabryelska and Marcin Sochal
Int. J. Mol. Sci. 2024, 25(17), 9427; https://doi.org/10.3390/ijms25179427 - 30 Aug 2024
Viewed by 578
Abstract
Obstructive Sleep Apnea (OSA) is a disorder characterized by repeated upper airway collapse during sleep, leading to apneas and/or hypopneas, with associated symptoms like intermittent hypoxia and sleep fragmentation. One of the agents contributing to OSA occurrence and development seems to be serotonin [...] Read more.
Obstructive Sleep Apnea (OSA) is a disorder characterized by repeated upper airway collapse during sleep, leading to apneas and/or hypopneas, with associated symptoms like intermittent hypoxia and sleep fragmentation. One of the agents contributing to OSA occurrence and development seems to be serotonin (5-HT). Currently, the research focuses on establishing and interlinking OSA pathogenesis and the severity of the disease on the molecular neurotransmitter omnipresent in the human body—serotonin, its pathway, products, receptors, drugs affecting the levels of serotonin, or genetic predisposition. The 5-HT system is associated with numerous physiological processes such as digestion, circulation, sleep, respiration, and muscle tone—all of which are considered factors promoting and influencing the course of OSA because of correlations with comorbid conditions. Comorbidities include obesity, physiological and behavioral disorders as well as cardiovascular diseases. Additionally, both serotonin imbalance and OSA are connected with psychiatric comorbidities, such as depression, anxiety, or cognitive dysfunction. Pharmacological agents that target 5-HT receptors have shown varying degrees of efficacy in reducing the Apnea-Hypopnea Index and improving OSA symptoms. The potential role of the 5-HT signaling pathway in modulating OSA provides a promising avenue for new therapeutic interventions that could accompany the primary treatment of OSA—continuous positive airway pressure. Thus, this review aims to elucidate the complex role of 5-HT and its regulatory mechanisms in OSA pathophysiology, evaluating its potential as a therapeutic target. We also summarize the relationship between 5-HT signaling and various physiological functions, as well as its correlations with comorbid conditions. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>A graphical representation of the associations between 5-HT psychiatric comorbidities—specifically anxiety disorder and depression—within the context of OSA. This aims to highlight the prevalence, the benefits of CPAP usage, and the role of 5-HT neurotransmission. Abbreviations: continuous positive airway pressure (CPAP), obstructive sleep apnea (OSA), healthy controls (HC), serotonin (5-HT).</p>
Full article ">Figure 2
<p>Graphical representation of associations between the 5-HT and psychiatric comorbidities being cognitive impairment within the context of OSA. This aims to highlight the risk factors for cognitive impairment, a consequence of OSA being intermittent hypoxia, and its further implications. Abbreviations: obstructive sleep apnea (OSA), serotonin (5-HT).</p>
Full article ">
22 pages, 1158 KiB  
Article
How Does the Psychological Impact of COVID-19 Affect the Management Strategies of Individuals with Type 1 and Type 2 Diabetes? A Mixed-Method Study
by Norah Abdullah Bazek Madkhali
Healthcare 2024, 12(17), 1710; https://doi.org/10.3390/healthcare12171710 - 27 Aug 2024
Viewed by 512
Abstract
(1) Background: During and after the pandemic, individuals with type 1 and type 2 diabetes struggled to maintain a healthy lifestyle due to psychological distress and the struggle to accommodate contextual challenges and changes in their family and work obligations and expectations. This [...] Read more.
(1) Background: During and after the pandemic, individuals with type 1 and type 2 diabetes struggled to maintain a healthy lifestyle due to psychological distress and the struggle to accommodate contextual challenges and changes in their family and work obligations and expectations. This study aims to explore the long-term impacts of the pandemic on proactive self-management behaviors and outcomes that consider contextual and environmental factors, such as family and work dynamics. (2) Methods: In this mixed-method study, data were collected from 418 participants using the Hospital Anxiety and Depression Scale (HADS) and the Insomnia Severity Index (ISI), followed by 16 individual interviews. (3) Results: The prevalence of depression was 37.1%, that of anxiety was 59.1%, and that of insomnia was 66.3%. Significant differences were observed in anxiety by age (p = 0.02), while individuals with other comorbidities were more likely to report insomnia (p = 0.3). Overall, various challenges during the pandemic have exacerbated emotional distress and complicated self-care routines and adherence to healthy lifestyles. (5) Conclusions: The COVID-19 pandemic has prompted individuals with type 1 and 2 diabetes to adopt alternative health-management methods, such as self-care, proactive initiatives, and daily challenges. Enhancing proactiveness, awareness, and an understanding of individuals’ needs is crucial for alleviating stress, controlling disease, and preparing for potential future health crises in the wake of the pandemic’s long-term effects. Full article
Show Figures

Figure 1

Figure 1
<p>Flow diagram of the recruitment of the participants.</p>
Full article ">Figure 2
<p>Psychological distress, contextual challenges, and proactive self-management experiences among individuals with diabetes during and after the COVID-19 pandemic.</p>
Full article ">
19 pages, 5595 KiB  
Article
Effects of DNA Methylation of HPA-Axis Genes of F1 Juvenile Induced by Maternal Density Stress on Behavior and Immune Traits in Root Voles (Microtus oeconomus)—A Field Experiment
by Shouyang Du, Guozhen Shang, Xin Tian, Zihan Liu, Yanbin Yang, Hongxing Niu, Jianghui Bian, Yan Wu and Jinyou Ma
Animals 2024, 14(17), 2467; https://doi.org/10.3390/ani14172467 - 25 Aug 2024
Viewed by 485
Abstract
The literature shows that maternal stress can influence behavior and immune function in F1. Yet, most studies on these are from the laboratory, and replicated studies on the mechanisms by which maternal stress drives individual characteristics are still not fully understood in wild [...] Read more.
The literature shows that maternal stress can influence behavior and immune function in F1. Yet, most studies on these are from the laboratory, and replicated studies on the mechanisms by which maternal stress drives individual characteristics are still not fully understood in wild animals. We manipulated high- and low-density parental population density using large-scale field enclosures and examined behavior and immune traits. Within the field enclosures, we assessed anti-keyhole limpet hemocyanin immunoglobulin G (anti-KLH IgG) level, phytohemagglutinin (PHA) responses, hematology, cytokines, the depressive and anxiety-like behaviors and prevalence and intensity of coccidial infection. We then collected brain tissue from juvenile voles born at high or low density, quantified mRNA and protein expression of corticotropin-releasing hormone (CRH) and glucocorticoid receptor gene (NR3C1) and measured DNA methylation at CpG sites in a region that was highly conserved with the prairie vole CRH and NR3C1 promoter. At high density, we found that the F1 had a lower DNA methylation level of CRH and a higher DNA methylation level of NR3C1, which resulted in an increase in the expression levels of the CRH mRNA and protein expression and further reduced the expression levels of the NR3C1 mRNA and protein expression, and ultimately led to have delayed responses to acute immobilization stress. Juvenile voles born at high density also reduced anti-KLH IgG levels and PHA responses, increased cytokines, and depressive and anxiety-like behaviors, and the effects further led to higher coccidial infection. From the perspective of population density inducing the changes in behavior and immunity at the brain level, our results showed a physiological epigenetic mechanism for population self-regulation in voles. Our results indicate that altering the prenatal intrinsic stress environment can fundamentally impact behavior and immunity by DNA methylation of HPA-axis genes and can further drive population fluctuations in wild animals. Full article
Show Figures

Figure 1

Figure 1
<p>Fecal corticosterone metabolite (FCM) levels across live-trapping sessions in high- and low-density F1. Data were expressed as mean ± SE for each group. “*”, <span class="html-italic">p</span> &lt; 0.05 vs. the low-density group. N = 93 and 117 for FCM measurements in low-density and high-density F1 group, respectively.</p>
Full article ">Figure 2
<p>Fecal corticosterone metabolite (FCM) response to acute immobilization stress between two replicate treatment male (<b>a</b>) (mean ± SE) (N = 19 and 38 in the low-density and high-density F1 group, respectively) and female (<b>b</b>) (mean ± SE) (N = 22 and 36 in the low-density and high-density F1 group, respectively) F1 groups. <span class="html-italic">p</span> &lt; 0.05 vs. basal level for the low-density (hash symbol) and high-density groups (asterisk).</p>
Full article ">Figure 3
<p>DNA methylation levels of corticotropin-releasing hormone (CRH) gene (<b>a</b>) (mean ± SE) and glucocorticoid receptor gene (<span class="html-italic">NR3C1</span>) (<b>b</b>) (mean ± SE). “**” indicates <span class="html-italic">p</span> &lt; 0.01 between low density and high density.</p>
Full article ">Figure 4
<p>The mRNA expression of corticotropin-releasing hormone (CRH) gene (<b>a</b>) (mean ± SE) and glucocorticoid receptor gene (<span class="html-italic">NR3C1</span>) (<b>b</b>) (mean ± SE). “**” and “***” indicate <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001 between low density and high density, respectively.</p>
Full article ">Figure 5
<p>The protein expression of corticotropin-releasing hormone (CRH) (<b>a</b>) and glucocorticoid receptor gene (<span class="html-italic">NR3C1</span>) (<b>b</b>) in F1. Data were expressed as mean ± SE for each group. “*”, “**” and “***” indicate <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 between low density and high density, respectively.</p>
Full article ">Figure 6
<p>The activity trajectory and activity of root voles in the open field (<b>a</b>–<b>e</b>) (mean ± SE) (male: N = 20 and 27, female: N = 25 and 29, in the low-density and high-density F1 group, respectively) and the elevated plus maze (<b>f</b>–<b>j</b>) (mean ± SE) (male: N = 25 and 28, female: N = 19 and 27, in the low-density and high-density F1 group, respectively). “*” and “***” indicate <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.001 between low density and high density, respectively.</p>
Full article ">Figure 7
<p>The levels of IL-1β (<b>a</b>) (mean ± SE), IL-6 (<b>b</b>) (mean ± SE), TNF-α (<b>c</b>) (mean ± SE), and IL-10 (<b>d</b>) (mean ± SE) between two replicate treatment F1 groups. “*”, “**” and “***” indicate <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 between low density and high density, respectively. N = 26 and 32 for males, N = 18 and 22 for females, in the low-density and high-density F1 group, respectively.</p>
Full article ">Figure 8
<p>Phytohemagglutinin (PHA) response (<b>a</b>) (mean ± SE) (male: N = 23 and 33, female: N = 20 and 28, in the low−density and high−density F1 group, respectively) and anti−keyhole limpet hemocyanin immunoglobulin G (anti−KLH IgG) level (<b>b</b>) (mean ± SE) (male: N = 32 and 35, female: N = 19 and 23, in the low−density and high−density F1 group, respectively) between two replicate treatment F1 groups. “**” and “***” indicate <span class="html-italic">p</span> &lt; 0.01 and <span class="html-italic">p</span> &lt; 0.001 between low density and high density, respectively.</p>
Full article ">Figure 9
<p>Coccidial prevalence (<b>a</b>) (mean ± SE) and intensity of infection (<b>b</b>) (mean ± SE) in two replicate F1 groups. * Indicates <span class="html-italic">p</span> &lt; 0.05 between low density and high density. N = 20 and 33 for males, N = 15 and 14 for females, in the low-density and high-density F1 group, respectively.</p>
Full article ">
22 pages, 11784 KiB  
Article
An Analysis of the Intestinal Microbiome Combined with Metabolomics to Explore the Mechanism of How Jasmine Tea Improves Depression in CUMS-Treated Rats
by Yangbo Zhang, Yong Lin, Yifan Xiong, Jianan Huang and Zhonghua Liu
Foods 2024, 13(16), 2636; https://doi.org/10.3390/foods13162636 - 22 Aug 2024
Viewed by 543
Abstract
Recently, research has confirmed that jasmine tea may help improve the depressive symptoms that are associated with psychiatric disorders. Our team previously found that jasmine tea improved the depressive-like behavior that is induced by chronic unpredictable mild stress (CUMS) in Sprague Dawley (SD) [...] Read more.
Recently, research has confirmed that jasmine tea may help improve the depressive symptoms that are associated with psychiatric disorders. Our team previously found that jasmine tea improved the depressive-like behavior that is induced by chronic unpredictable mild stress (CUMS) in Sprague Dawley (SD) rats. We hypothesized that the metabolic disorder component of depression may be related to the gut microbiota, which may be reflected in the metabolome in plasma. The influence of jasmine tea on gut microbiota composition and the association with depressive-related indexes were explored. Furthermore, the metabolites in plasma that are related to the gut microbiota were identified. SD rats were treated with control or CUMS and administrated jasmine tea for 8 weeks. The 16S rRNA gene amplicon sequencing was used to analyze the gut microbiota in feces samples, and untargeted metabolomics was used to analyze the metabolites in plasma. The results found that jasmine tea significantly ameliorated the depressive behavior induced by CUMS, significantly improved the neurotransmitter concentration (BDNF and 5-HT), and decreased the pro-inflammation levels (TNF-α and NF-κB). The intervention of jasmine tea also alleviated the dysbiosis caused by CUMS; increased the relative abundance of Bacteroides, Blautia, Clostridium, and Lactobacillus; and decreased Ruminococcus and Butyrivibrio in the CUMS-treated rats. Furthermore, the serum metabolites of the CUMS-treated rats were reversed after the jasmine tea intervention, i.e., 22 were up-regulated and 18 were down-regulated, which may have a close relationship with glycerophospholipid metabolism pathways, glycine serine and threonine metabolism pathways, and nicotinate and nicotinamide metabolism pathways. Finally, there were 30 genera of gut microbiota related to the depressive-related indexes, and 30 metabolites in the plasma had a strong predictive ability for depressive behavior. Potentially, our research implies that the intervention of jasmine tea can ameliorate the depression induced by CUMS via controlling the gut flora and the host’s metabolism, which is an innovative approach for the prevention and management of depression. Full article
(This article belongs to the Section Foodomics)
Show Figures

Figure 1

Figure 1
<p>The schedule of experimental procedures. SPT, sugar preference test; OFT, open-field test; SFT, forced swimming test.</p>
Full article ">Figure 2
<p>Results of the behavioral tests. (<b>A</b>) Body weight during the experiment. (<b>B</b>) Body weight gain during the experiment. (<b>C</b>) Food intake during the experiment. (<b>D</b>) Sugar preference during the experiment. (<b>E</b>) Immobility time comparison of FST. (<b>F</b>) The comparison of number of inners, outers, and standing in OFT. Con, control; Mod, model; NF, CUMS treated with jasmine tea. SPT, sugar preference test; FST, force swimming test; OFT, open field test. # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 versus the control group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 versus the model group.</p>
Full article ">Figure 3
<p>The levels of neurotransmitter and inflammation factors among different tissues. (<b>A</b>) 5-HT; (<b>B</b>) BDNF; (<b>C</b>) TNF-α; (<b>D</b>) NF-κB. # <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; 0.01 versus the 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 versus the model group.</p>
Full article ">Figure 4
<p>The ameliorations of phenotype in hippocampus and colon of CUMS-induced rats after jasmine tea administration. (<b>A</b>) HE staining of the hippocampus. (<b>B</b>) Nissl staining of the hippocampus. (<b>C</b>) HE staining of the colon. →, the comparison of special structures in the figure.</p>
Full article ">Figure 5
<p>Gut microbial characteristics of Con, Mod, and NF. (<b>A</b>) Alpha-phylogenetic diversity analysis showed that depressive-like rats induced by CUMS had lower microbial richness in four indexes relative to controls; these indexes had increased when administrated with jasmine tea. (<b>B</b>) Principal component analysis (PCA) revealed that the gut microbiome composition in rats with depressive-like symptoms induced by CUMS was markedly distinct from that of Con, and the intestinal microbial composition of depressive-like induced by CUMS was restored after being treated with jasmine tea. (<b>C</b>) Venn diagram for taxonomy of gut microbes based on genus level. (<b>D</b>) Circus circle diagram of the top 10 relative abundances in intestinal microbial classification based on genus level. (<b>E</b>) The gut microbiota compositions among the experimental groups at the phylum level. (<b>F</b>) The relative abundance of gut microbes at the genus level. # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 versus the control group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, versus the model group.</p>
Full article ">Figure 6
<p>Linear discriminant analysis effect size (LEfSe) analysis was conducted with a threshold of LDA &gt; 2.0. The results were visualized using a cladogram (<b>A</b>) and a histogram (<b>B</b>) and showed 35 genera responsible for discriminating in Con, Mod, and NF.</p>
Full article ">Figure 7
<p>Jasmine tea intervention regulated the gut microbiota in CUMS-induced depressive rats. (<b>A</b>) Heatmap of the gut microbiome in genus levels. (<b>B</b>) Phylogenetic tree and heatmap of abundances distribution between groups in phylum level. (<b>C</b>) The comparison of relative abundance of gut microbial community members was conducted at the genus level among three groups. Data represent the mean ± SEM of six rats in each group. # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 versus the control group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 versus the model group.</p>
Full article ">Figure 8
<p>Associations of gut microbes with neurotransmitters and inflammation factors based on Spearman correlation analysis. (<b>A</b>) The composition of intestinal microorganisms based on the genus level and environmental factors being analyzed through a mapping test. (<b>B</b>) Overreplacement test map of intestinal microorganisms based on generic levels and environmental factors. (<b>C</b>) Visualization of Spearman’s rank correlation in the form of a heatmap of 30 genera and neurotransmitters, with inflammations in colon. * <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 ">Figure 9
<p>Multivariate analysis of metabolic profiles of Con, Mod, and NF. (<b>A</b>–<b>C</b>) The evaluation scatter plots of different groups from the PLS-DA data. (<b>D</b>) Con and Mod pairwise comparison OPLS-DA evaluation scatter plots. (<b>E</b>) Mod and NF pairwise comparison OPLS-DA evaluation scatter plots. (<b>G</b>) Stacked column chart of the percentage of metabolites that play a biological role. (<b>F</b>) Pathway topology enrichment among different treatments.</p>
Full article ">Figure 10
<p>Associations of gut microbes with neurotransmitters and inflammation factors based on Spearman correlation analysis. * <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 ">Figure 11
<p>Network diagram of associations of gut based on lefse-genus and metabolites based on OPLS-DA.</p>
Full article ">
16 pages, 17053 KiB  
Article
Polygonatum sibiricum Polysaccharides Alleviate Depressive-like Symptoms in Chronic Restraint Stress-Induced Mice via Microglial Regulation in Prefrontal Cortex
by Zhong-Yu Yuan, Xuan Zhang, Zong-Zhong Yu, Xin-Yu Wang, Zi-Heng Zeng, Meng-Xuan Wei, Meng-Ting Qiu, Jun Wang, Jie Cheng and Li-Tao Yi
Polymers 2024, 16(16), 2358; https://doi.org/10.3390/polym16162358 - 20 Aug 2024
Viewed by 412
Abstract
Microglia respond to stressors by secreting cytokines or growth factors, playing a crucial role in maintaining brain homeostasis. While the antidepressant-like effects of Polygonatum sibiricum polysaccharides (PSPs) have been observed in mice, their potential effectiveness involving microglial regulation remains unknown. This study investigates [...] Read more.
Microglia respond to stressors by secreting cytokines or growth factors, playing a crucial role in maintaining brain homeostasis. While the antidepressant-like effects of Polygonatum sibiricum polysaccharides (PSPs) have been observed in mice, their potential effectiveness involving microglial regulation remains unknown. This study investigates the antidepressant-like mechanism of PSP by regulating microglial phenotype and signaling pathways in the prefrontal cortex of chronic restraint stress (CRS)-induced mice. PSP was extracted, purified, characterized, and orally administered to CRS mice. High-performance gel permeation chromatography (HPGPC) revealed that PSP has a molecular weight of 5.6 kDa. Scanning electron microscopy (SEM) and atomic force microscopy (AFM) showed that PSP exhibited a layered structure with densely packed, irregular surfaces. PSP treatment significantly increased sucrose preference (low: 71%, p < 0.01; medium: 69%, p < 0.05; high: 75%, p < 0.001 vs. CRS: 58%) and reduced immobility time (low: 74 s, p < 0.01; medium: 68 s, p < 0.01; high: 79 s, p < 0.05 vs. CRS: 129 s), indicating the alleviation of depressive-like behaviors. PSP inhibited microglial activation (PSP, 131/mm2 vs. CRS, 173/mm2, p = 0.057), reversing CRS-induced microglial hypertrophy and hyper-ramification. Furthermore, PSP inactivated microglial activation by inhibiting NLRP3/ASC/caspase-1/IL-1β signaling pathways, increasing BDNF synthesis and activating brain-derived neurotrophic factor (BDNF)-mediated neurogenesis (PSP, 80/per DG vs. CRS, 49/per DG, p < 0.01). In conclusion, PSP exerts antidepressant-like effects through the regulation of microglial activity and neuroinflammatory pathways, indicating it as a potential natural compound for depression treatment. Full article
(This article belongs to the Special Issue Biopolymer-Based Materials in Medical Applications)
Show Figures

Figure 1

Figure 1
<p>Characterization of <span class="html-italic">Polygonatum sibiricum</span> polysaccharides (PSP). (<b>A</b>) Infrared spectrum of PSP. (<b>B</b>) High-performance gel permeation chromatography (HPGPC) analysis of PSP. (<b>C</b>) SEM image of PSP at 500× magnification. (<b>D</b>) SEM image of PSP at 5000× magnification. (<b>E</b>) Two-dimensional morphology of AFM. (<b>F</b>) Three-dimensional morphology of AFM.</p>
Full article ">Figure 2
<p>Behavioral assessments of mice treated with PSP in a CRS model (<span class="html-italic">n</span> = 11). (<b>A</b>) Sucrose preference, as anhedonia symptom of depression was reversed by PSP. (<b>B</b>) Immobility time, as despair symptom of depression was attenuated by PSP. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. normal group. * <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 vs. CRS group.</p>
Full article ">Figure 3
<p>PSP partly inactivated TLR4/NF-κB signaling and significantly inhibited NLRP3/ASC/caspase-1/IL-1β signaling pathway in the prefrontal cortex of CRS-induced mice (<span class="html-italic">n</span> = 6). (<b>A</b>) TLR4. (<b>B</b>) pNF-κB/NF-κB ratio. (<b>C</b>) NLRP3. (<b>D</b>) ASC. (<b>E</b>) Cleaved caspase-1. (<b>F</b>) Pro IL-1β. (<b>G</b>) Mature IL-1β. N: normal; C: CRS; F: fluoxetine; L: 100 mg/kg PSP; M: 200 mg/kg PSP; H: 400 mg/kg PSP. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. normal group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. CRS group.</p>
Full article ">Figure 4
<p>PSP attenuated microglial activation in the prefrontal cortex of CRS-induced mice (<span class="html-italic">n</span> = 5). (<b>A</b>) Immunofluorescence staining of microglia: representative images showing immunofluorescence staining of microglia in the prefrontal cortex. Blue indicates DAPI-stained nuclei, and red indicates Iba-1-stained microglia. Scale bar: 50 μm. (<b>B</b>) Quantification of microglial number per square millimeter. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. normal group.</p>
Full article ">Figure 5
<p>PSP treatment reversed CRS-induced prefrontal cortex microglial hypertrophy and hyper-ramification (<span class="html-italic">n</span> = 5). (<b>A</b>) Representative 3D reconstructions of microglia show increased size and complexity in the CRS group compared to the control, with PSP treatment reducing these changes. Scale bar: 5 μm. (<b>B</b>–<b>J</b>) Quantitative analysis: CRS significantly increased microglial area, length, volume, full branch depth, branch points, and Sholl intersections. PSP treatment significantly reduced area, volume, and Sholl intersections, with trends towards normalization in other parameters. These results suggest that PSP mitigates CRS-induced microglial hypertrophy and hyper-ramification, restoring more normal morphology. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. normal group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. CRS group.</p>
Full article ">Figure 6
<p>Effects of PSP on inflammatory marker expression in prefrontal cortex microglia of CRS-induced mice. (<b>A</b>) Representative immunofluorescence staining images of TLR4 (green) expression in microglia (Iba-1, red) with DAPI (blue) staining. (<b>B</b>) Quantification shows increased TLR4 expression in the CRS group reduced by PSP treatment. (<b>C</b>) NLRP3 (green) expression images. (<b>D</b>) Quantification shows increased NLRP3 in the CRS group, reduced by PSP. (<b>E</b>) ASC (green) expression images. (<b>F</b>) Quantification shows increased ASC in the CRS group, reduced by PSP. (<b>G</b>) IL-1β (green) expression images. (<b>H</b>) Quantification shows a significant increase in IL-1β in the CRS group, significantly reduced by PSP. Scale bar: 4 μm. Within Iba-1 means the signal of the target protein was merged by the signal of the Iba-1 constructed surface by Imaris v9.0.1. Merge means blue, red, and green signals overlaying. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. normal group. * <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 vs. CRS group.</p>
Full article ">Figure 7
<p>PSP partly increased microglial BDNF expression in the prefrontal cortex and enhanced neurogenesis in the DG of CRS-induced mice. (<b>A</b>) Representative immunofluorescence staining, BDNF (green) in microglia (Iba-1, red) with DAPI-stained nuclei (blue). (<b>B</b>) Quantification showing a significant decrease in relative BDNF fluorescence intensity in the CRS group that is partially reversed by PSP. (<b>C</b>) Representative immunofluorescence staining presents DCX staining to visualize newborn neurons in the DG. (<b>D</b>) Quantification showing the number of newborn neurons per DG. Within Iba-1 means the signal of the target protein was merged by the signal of the Iba-1 constructed surface by Imaris. Merge means blue, red, and green signals overlaying. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. normal group. ** <span class="html-italic">p</span> &lt; 0.01 vs. CRS group.</p>
Full article ">
22 pages, 2403 KiB  
Review
Antidepressant- and Anxiolytic-like Effects of Pomegranate: Is It Acting by Common or Well-Known Mechanisms of Action?
by Erika Estrada-Camerena, Carolina López-Rubalcava, Nelly Maritza Vega-Rivera and María Eva González-Trujano
Plants 2024, 13(16), 2205; https://doi.org/10.3390/plants13162205 - 9 Aug 2024
Viewed by 763
Abstract
The pharmacological effects of pomegranates have been described considering metabolic aspects such as hypoglycemic and hypolipidemic activities. The pomegranate extract has activity on the central nervous system (CNS) as a natural antidepressant and anxiolytic. The chemical composition of pomegranates is complex since the [...] Read more.
The pharmacological effects of pomegranates have been described considering metabolic aspects such as hypoglycemic and hypolipidemic activities. The pomegranate extract has activity on the central nervous system (CNS) as a natural antidepressant and anxiolytic. The chemical composition of pomegranates is complex since the bioactive compounds are multiple secondary metabolites that have been identified in the extracts derived from the peel, seed, flowers, leaves, or in their combination; so, it has not been easy to identify an individual compound as responsible for its observed pharmacological properties. From this point of view, the present review analyzes the effects of crude extracts or fractions of pomegranates and their possible mechanisms of action concerning antidepressant- and anxiolytic-like effects in animal models. Serotonin receptors, estrogen receptors, the peroxisome proliferator-activated receptor gamma (PPARγ), or monoamine oxidase enzymes, as well as potent antioxidant and neuroplasticity properties, have been described as possible mediators involved in the antidepressant- and anxiolytic-like behaviors after pomegranate treatment. The pharmacological effects observed on the CNS in experimental models associated with a specific stress level suggest that pomegranates could simultaneously modulate the stress response by activating several targets. For the present review, scientific evidence was gathered to integrate it and suggest a possible pathway for mediators to be involved in the mechanisms of action of the pomegranate’s antidepressant- and anxiolytic-like effects. Furthermore, the potential benefits are discussed on comorbid conditions with anxiety and depression, such as perimenopause transition and pain. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of phytoestrogen classification. Pomegranate and berries (strawberries, raspberries, blueberries, and blackberries) and nuts (walnuts, pecans, and chestnuts) are primary sources of <span class="underline">ellagitannins</span>, which are biotransformed in urolithins by microbiota (1); soy-derived products, groundnut, and tea are the primary source of <span class="underline">isoflavones</span>, which are finally converted to genistein and daidzein and, finally, to equol after a biotransformation process (2). <span class="underline">Lignans </span>(matairesinol and secoisolariciresinol) come from cereals (barley, oats, wheat, and corn), vegetables (curly kale, broccoli, asparagus, carrots, and garlic), fruits (apricot, strawberry, peach, and orange), and nuts (flaxseed, sesame, sunflower, and cashew) and are biotransformed to enterolactone and enterodiol (3). <span class="underline">Stilbens</span> represented by resveratrol are found in wine, peanut, red grapes, cocoa, pistachios, and blueberries (4). <span class="underline">Coumestans,</span> represented by coumestrol, can be found in legumes, sprouts of soy, alfalfa, Brussels sprouts, clover, and chickpeas (5). Stilbens and coumestans production appears to be independent of microbiota [<a href="#B16-plants-13-02205" class="html-bibr">16</a>,<a href="#B27-plants-13-02205" class="html-bibr">27</a>,<a href="#B28-plants-13-02205" class="html-bibr">28</a>].</p>
Full article ">Figure 2
<p>Schematic representation of the most representative groups of polyphenols classified according to their chemical structure in flavonoids (flavan nucleus), no flavonoids (phenolic amides and acids), and other phenolic groups (lignans, stilbenes, and hydrolyzed tannins) [<a href="#B21-plants-13-02205" class="html-bibr">21</a>,<a href="#B22-plants-13-02205" class="html-bibr">22</a>,<a href="#B29-plants-13-02205" class="html-bibr">29</a>,<a href="#B30-plants-13-02205" class="html-bibr">30</a>].</p>
Full article ">Figure 3
<p>Comorbidity among depression, anxiety, and pain. These pathologies are the target of different active compounds from pomegranates, suggesting that their regular consumption could contribute to decreasing their symptomatology and improving these health conditions.</p>
Full article ">Figure 4
<p>A common mechanism of action for antidepressant, anxiolytic, and anti-inflammatory actions of the aqueous pomegranate extract (<span class="html-italic">Punicagranatum</span>). The bioactive compounds contained in pomegranate extract could increase the activity of monoamine oxidase type A (MAO-A) enzyme in serotonergic neurons, facilitating the levels of 5-hydroxi-indolacetic acid, which, in turn, activates the peroxisome proliferator-activated receptor gamma (PPARγ). Activation of this receptor in specific brain areas may contribute, via ERK1/2phosphorylation, to decreasing anxiety and reducing oxidative stress by decreasing reactive oxygen species formation (ROS). This could also impact the decrease in pro-inflammatory cytokines, which may influence inflammation and pain. In addition, pomegranate components can also activate estrogenic receptor type β, favoring the activity of the serotonergic system to exert antidepressant-like action and contribute to the activation of PPARγ.</p>
Full article ">
14 pages, 2161 KiB  
Article
Brain Region-Specific Expression Levels of Synuclein Genes in an Acid Sphingomyelinase Knockout Mouse Model: Correlation with Depression-/Anxiety-Like Behavior and Locomotor Activity in the Absence of Genotypic Variation
by Razvan-Marius Brazdis, Iulia Zoicas, Johannes Kornhuber and Christiane Mühle
Int. J. Mol. Sci. 2024, 25(16), 8685; https://doi.org/10.3390/ijms25168685 - 9 Aug 2024
Viewed by 604
Abstract
Accumulating evidence suggests an involvement of sphingolipids, vital components of cell membranes and regulators of cellular processes, in the pathophysiology of both Parkinson’s disease and major depressive disorder, indicating a potential common pathway in these neuropsychiatric conditions. Based on this interaction of sphingolipids [...] Read more.
Accumulating evidence suggests an involvement of sphingolipids, vital components of cell membranes and regulators of cellular processes, in the pathophysiology of both Parkinson’s disease and major depressive disorder, indicating a potential common pathway in these neuropsychiatric conditions. Based on this interaction of sphingolipids and synuclein proteins, we explored the gene expression patterns of α-, β-, and γ-synuclein in a knockout mouse model deficient for acid sphingomyelinase (ASM), an enzyme catalyzing the hydrolysis of sphingomyelin to ceramide, and studied associations with behavioral parameters. Normalized Snca, Sncb, and Sncg gene expression was determined by quantitative PCR in twelve brain regions of sex-mixed homozygous (ASM−/−, n = 7) and heterozygous (ASM+/−, n = 7) ASM-deficient mice, along with wild-type controls (ASM+/+, n = 5). The expression of all three synuclein genes was brain region-specific but independent of ASM genotype, with β-synuclein showing overall higher levels and the least variation. Moreover, we discovered correlations of gene expression levels between brain regions and depression- and anxiety-like behavior and locomotor activity, such as a positive association between Snca mRNA levels and locomotion. Our results suggest that the analysis of synuclein genes could be valuable in identifying biomarkers and comprehending the common pathological mechanisms underlying various neuropsychiatric disorders. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Brain-specific variation of α, β-, and γ-synuclein gene expression in twelve regions with uniformity across the three acid sphingomyelinase (ASM) genotypes. (<b>a</b>) <span class="html-italic">Snca</span>, (<b>b</b>) <span class="html-italic">Sncb</span>, and (<b>c</b>) <span class="html-italic">Sncg</span> were expressed differently in twelve brain regions: frontal cortex (FC), dorsal striatum (DS), lateral septum (LS), ventral striatum (VS), amygdala (AM), dorsal hippocampus (DH), thalamus (TH), hypothalamus (HY), ventral hippocampus (VH), dorsal mesencephalon (DM), ventral mesencephalon (VM), and cerebellum (CE). No statistically significant differences were observed between homozygous ASM-deficient (ASM−/−, <span class="html-italic">n</span> = 7), heterozygous ASM-deficient (ASM+/−, <span class="html-italic">n</span> = 7), and wild-type (ASM+/+, <span class="html-italic">n</span> = 5) mice. Data represent individual data points with means as bars.</p>
Full article ">Figure 2
<p>Heat maps of Spearman correlation coefficient (ρ) between (<b>a</b>) <span class="html-italic">Snca</span>, (<b>b</b>) <span class="html-italic">Sncb</span>, and (<b>c</b>) <span class="html-italic">Sncg</span> expression in twelve brain regions, frontal cortex (FC), dorsal striatum (DS), lateral septum (LS), ventral striatum (VS), amygdala (AM), dorsal hippocampus (DH), thalamus (TH), hypothalamus (HY), ventral hippocampus (VH), dorsal mesencephalon (DM), ventral mesencephalon (VM), and cerebellum (CE), for the entire group of mice (total, <span class="html-italic">n</span> = 19). ρ index ranges from −1 to +1; blue indicates a positive correlation, and red a negative correlation (darker color indicates a stronger correlation); white (ρ = 0) represents no correlation. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 for the significance level of the correlation.</p>
Full article ">Figure 3
<p>The behavioral phenotype of homozygous ASM knockout (ASM−/−, <span class="html-italic">n</span> = 7), heterozygous ASM-deficient (ASM+/−, <span class="html-italic">n</span> = 7), and wild-type (ASM+/+, <span class="html-italic">n</span> = 5) mice. (<b>a</b>) Percentage of immobility time, as an indicator of depression-like behavior, was assessed in the forced swim test; (<b>b</b>) Percentage of time spent in the open arms of the elevated plus-maze is an indicator of anxiety-like behavior; (<b>c</b>) The number of entries into the closed arm of the elevated plus-maze is an indicator of locomotor activity; (<b>a</b>,<b>b</b>) ASM−/− mice showed a reduced depression-like phenotype, but increased anxiety-like behavior compared with ASM+/+ mice. Locomotor activity was reduced in ASM−/− mice compared with ASM+/− mice. Data represent the means + SEM. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Heat maps of Spearman correlation coefficient (ρ) between (<b>a</b>) <span class="html-italic">Snca</span>, (<b>b</b>) <span class="html-italic">Sncb</span>, and (<b>c</b>) <span class="html-italic">Sncg</span> expression and depression-like behavior (D) expressed as percentage immobility in the forced swim test, anxiety-like behavior (A) indicated by the percentage of time spent in the open arms of the elevated plus-maze, and locomotor activity (L) assessed by the number of closed arm entries in the elevated plus-maze in twelve brain regions: frontal cortex (FC), dorsal striatum (DS), lateral septum (LS), ventral striatum (VS), amygdala (AM), dorsal hippocampus (DH), thalamus (TH), hypothalamus (HY), ventral hippocampus (VH), dorsal mesencephalon (DM), ventral mesencephalon (VM), and cerebellum (CE), for the entire group of mice [<span class="html-italic">n</span> = 19, male <span class="html-italic">n</span> = 8, female <span class="html-italic">n</span> = 11; wild-type (ASM+/+) <span class="html-italic">n</span> = 5, homozygous ASM-deficient (ASM−/−) <span class="html-italic">n</span> = 7 and heterozygous ASM-deficient (ASM+/−) <span class="html-italic">n</span> = 7]. ρ index ranges from −1 to +1; blue indicates a positive correlation, and red a negative correlation (darker color indicates a stronger correlation); white (ρ = 0) represents no correlation. * <span class="html-italic">p</span> &lt; 0.05 for the significance level of the correlation.</p>
Full article ">Figure 5
<p>Associations of synuclein expression data with behavioral measures: (<b>a</b>) Negative correlation of <span class="html-italic">Sncb</span> expression with depression-like behavior, expressed as percentage immobility in the forced swim test, in the ventral striatum (VS) of female (red, <span class="html-italic">n</span> = 8) and male (blue, <span class="html-italic">n</span> = 11) mice; (<b>b</b>) Positive correlation of <span class="html-italic">Snca</span> expression with percentage of time spent in the open arms of the elevated plus-maze, as an inverse indicator of anxiety-like behavior, in the amygdala (AM) of female (red) and male (blue) heterozygous ASM-deficient (ASM+/−, <span class="html-italic">n</span> = 7) mice; (<b>c</b>) Positive correlation of <span class="html-italic">Snca</span> expression with number of closed arm entries in the elevated plus-maze, as an indicator of locomotor activity, in the cerebellum (CE) of combined female homozygous ASM-deficient (ASM−/−, <span class="html-italic">n</span> = 1), heterozygous ASM-deficient (ASM+/−, <span class="html-italic">n</span> = 4), and wild-type (ASM+/+, <span class="html-italic">n</span> = 3) mice, (<b>d</b>) as well as in female ASM+/+ (<span class="html-italic">n</span> = 3) and male ASM+/+ (<span class="html-italic">n</span> = 2) mice. Linear regression line for the combined group with 95% confidence interval and statistics (Spearman correlation, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
13 pages, 547 KiB  
Article
The Roles of Affective Lability, Boredom, and Mindfulness in Predicting Number of Sex Partners within Women
by Carolyn A. Lorenzi, David C. de Jong and Rachel S. Faulkenberry
Sexes 2024, 5(3), 262-274; https://doi.org/10.3390/sexes5030020 - 5 Aug 2024
Viewed by 490
Abstract
Affective lability, a trait related to borderline personality disorder, bipolar disorder, eating disorders, and post-traumatic stress disorder, is associated with a higher number of lifetime sex partners. Among individuals who are affectively labile, boredom proneness, which has been linked to impulsive and risky [...] Read more.
Affective lability, a trait related to borderline personality disorder, bipolar disorder, eating disorders, and post-traumatic stress disorder, is associated with a higher number of lifetime sex partners. Among individuals who are affectively labile, boredom proneness, which has been linked to impulsive and risky sexual behaviors, might increase the likelihood of having more sex partners. Conversely, mindfulness has been found to be associated with healthy emotion regulation and lower impulsivity, and may enable a greater tolerance of affective lability and boredom, and, in turn, lower the sense of urgency to engage in sex to cope. Thus, the present study investigated the links between affective lability, boredom proneness, mindfulness, and number of sex partners in the last year. We predicted that affective lability would be positively associated with number of sex partners, and that this association would be moderated by boredom proneness and mindfulness in a three-way interaction. Adult women (N = 469, Mage = 25.15 years) were recruited from online communities and completed measures of affective lability, boredom proneness, trait mindfulness, and number of sex partners in the last year. None of the preregistered three-way interactions were supported; however, exploratory analyses revealed that, among women who reported rapid changes between depression and elation, those who were less likely to observe thoughts and sensations had more sex partners in the last year. Clinicians and researchers should further investigate which facets of mindfulness may protect against a higher number of sex partners in affectively labile individuals. Full article
Show Figures

Figure 1

Figure 1
<p>Simple slopes for predicting number of sex partners from Depression/Elation and Observe.</p>
Full article ">
24 pages, 2752 KiB  
Article
Design, Synthesis, and Biological Evaluations of Novel Thiazolo[4,5-d]pyrimidine Corticotropin Releasing Factor (CRF) Receptor Antagonists as Potential Treatments for Stress Related Disorders and Congenital Adrenal Hyperplasia (CAH)
by Md Rabiul Islam, Christos Markatos, Ioannis Pirmettis, Minas Papadopoulos, Vlasios Karageorgos, George Liapakis and Hesham Fahmy
Molecules 2024, 29(15), 3647; https://doi.org/10.3390/molecules29153647 - 1 Aug 2024
Viewed by 774
Abstract
Corticotropin-releasing factor (CRF) is a key neuropeptide hormone that is secreted from the hypothalamus. It is the master hormone of the HPA axis, which orchestrates the physiological and behavioral responses to stress. Many disorders, including anxiety, depression, addiction relapse, and others, are related [...] Read more.
Corticotropin-releasing factor (CRF) is a key neuropeptide hormone that is secreted from the hypothalamus. It is the master hormone of the HPA axis, which orchestrates the physiological and behavioral responses to stress. Many disorders, including anxiety, depression, addiction relapse, and others, are related to over-activation of this system. Thus, new molecules that may interfere with CRF receptor binding may be of value to treat neuropsychiatric stress-related disorders. Also, CRF1R antagonists have recently emerged as potential treatment options for congenital adrenal hyperplasia. Previously, several series of CRF1 receptor antagonists were developed by our group. In continuation of our efforts in this direction, herein we report the synthesis and biological evaluation of a new series of CRF1R antagonists. Representative compounds were evaluated for their binding affinities compared to antalarmin. Four compounds (2, 5, 20, and 21) showed log IC50 values of −8.22, −7.95, −8.04, and −7.88, respectively, compared to −7.78 for antalarmin. This result indicates that these four compounds are superior to antalarmin by 2.5, 1.4, 1.7, and 1.25 times, respectively. It is worth mentioning that compound 2, in terms of IC50, is among the best CRF1R antagonists ever developed in the last 40 years. The in silico physicochemical properties of the lead compounds showed good drug-like properties. Thus, further research in this direction may lead to better and safer CRF receptor antagonists that may have clinical applications, particularly for stress-related disorders and the treatment of congenital adrenal hyperplasia. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Important CRF<sub>1</sub>R antagonists.</p>
Full article ">Figure 2
<p>Structure of Tildacerfont and Crinecerfont.</p>
Full article ">Figure 3
<p>General structure of synthesized analogs.</p>
Full article ">Figure 4
<p>General synthetic scheme for intermediate and final target compounds.</p>
Full article ">Figure 5
<p>% inhibition of [<sup>125</sup>I]-Try<sup>0</sup> sauvagine-specific binding by 100 nM of test compounds on membranes from HEK293 stably expressing human CRF<sub>1</sub> receptors. In the absence of the test compound, the inhibition is 0%.</p>
Full article ">Figure 6
<p>Competitive binding isotherms of compounds <b>2</b>, <b>5</b>, and <b>14</b> (<b>a</b>), compounds <b>10</b> and <b>23</b> (<b>b</b>), compounds <b>19</b> and <b>20</b> (<b>c</b>) and <b>21</b> and <b>22</b> (<b>d</b>) to human CRF<sub>1</sub> receptors. Antalarmin was used as a standard drug.</p>
Full article ">Figure 6 Cont.
<p>Competitive binding isotherms of compounds <b>2</b>, <b>5</b>, and <b>14</b> (<b>a</b>), compounds <b>10</b> and <b>23</b> (<b>b</b>), compounds <b>19</b> and <b>20</b> (<b>c</b>) and <b>21</b> and <b>22</b> (<b>d</b>) to human CRF<sub>1</sub> receptors. Antalarmin was used as a standard drug.</p>
Full article ">Figure 7
<p>Structure of compound <b>11</b> with more than four carbons at C-7 versus other two compounds (<b>10</b> and <b>13</b>) with 3–4 carbons in the side chain at C-7 position.</p>
Full article ">Figure 8
<p>Representative compounds with different substituted phenyl groups at the N-3 position.</p>
Full article ">
18 pages, 3400 KiB  
Article
Antidepressant-like and Beneficial Effects of a Neoponcirin-Beta-Cyclodextrin Inclusion Complex in Mice Exposed to Prolonged Stress
by Luis José López Méndez, Lucía Martínez-Mota, Julia Cassani, Lilian Mayagoitia-Novales, Gloria Benítez-King, Luis Enrique Becerril-Villanueva, Ana María Dorantes-Barrón, Noé Jurado-Hernández and Rosa Estrada-Reyes
Int. J. Mol. Sci. 2024, 25(15), 8289; https://doi.org/10.3390/ijms25158289 - 29 Jul 2024
Viewed by 582
Abstract
Neoponcirin causes anxiolytic-like effects in mice when administered intraperitoneally but not orally. Neoponcirin is non-water-soluble and insoluble in solvents, and in medium acid, it isomerizes, reducing its bioavailability. To improve the pharmacological properties of neoponcirin, we formed a neoponcirin complex with beta-cyclodextrin (NEO/βCD), [...] Read more.
Neoponcirin causes anxiolytic-like effects in mice when administered intraperitoneally but not orally. Neoponcirin is non-water-soluble and insoluble in solvents, and in medium acid, it isomerizes, reducing its bioavailability. To improve the pharmacological properties of neoponcirin, we formed a neoponcirin complex with beta-cyclodextrin (NEO/βCD), which was characterized by FT-IR, UV-Vis, and NMR, and their solubility profile. We evaluated the antidepressant-like effects of NEO/βCD acutely administered to mice orally in the behavioral paradigms, the tail suspension (TST) and the forced swimming (FST) tests. We also analyzed the benefits of repeated oral doses of NEO/βCD on depression- and anxiety-like behaviors induced in mice by chronic unpredictable mild stress (CUMS), using the FST, hole board, and open field tests. We determined the stressed mice’s expression of stress-related inflammatory cytokines (IL-1β, IL-6, and TNFα) and corticosterone. Results showed that a single or chronic oral administration of NEO/βCD caused a robust antidepressant-like effect without affecting the ambulatory activity. In mice under CUMS, NEO/βCD also produced anxiolytic-like effects and avoided increased corticosterone and IL-1β levels. The effects of the NEO/βCD complex were robust in both the acute and the stress chronic models, improving brain neurochemistry and recovering immune responses previously affected by prolonged stress. Full article
Show Figures

Figure 1

Figure 1
<p>Comparison of <sup>1</sup>H NMR spectra profile displacement of NEO/βCD (neoponcirin/beta cyclodextrin Inclusion Complex; IC) concerning NEO (neoponcirin). Drugs were dissolved in DMSO-d6. (<b>A</b>) <span style="color:#7030A0">---NEO free</span>, (<b>B</b>) <span style="color:#70AD47">---IC complex</span>, and (<b>C</b>) <span style="color:#2F5496">---βCD free</span>.</p>
Full article ">Figure 2
<p>UV-Vis absorption spectra of NEO/βCD in comparison with NEO (<b>A</b>), FT-IR spectra of NEO (purple), βCD (black), and βCD/NEO complex (green) (<b>B</b>).</p>
Full article ">Figure 3
<p>Drug dissolution profiles of NEO and the NEO/βCD complex. The points represent the average over three repeats. Remarkably, the cumulative dissolution of NEO reached 12.93% ± 4.506%, whereas the dissolution for NEO/βCD notably exceeded this at 79.74% ± 3.619%. These findings unequivocally highlight a significant increase in the complex’s solubility and dissolution rate concerning the NEO.</p>
Full article ">Figure 4
<p>Effects of a single oral dose of NEO/βCD in the tail suspension (<b>A</b>) and the forced swimming (<b>B</b>) tests. A, NEO/βCD, CTL, control group; CMI 25, clomipramine at 25 mg/kg; and NEO/βCD, complex at different tail suspension test (TST) doses.B control group; CMI 25, clomipramine at 25 mg/kg; NEO/βCD, complex at different doses in the forced swimming test (FST). Data represent the mean ± standard error of the mean of the accumulated immobility time of groups of 8 to 10 mice each. Differences between treatments were analyzed with non-parametric Kruskal–Wallis’ one-way ANOVA (*** <span class="html-italic">p</span> ≤ 0.001) and pairwise multiple comparison procedures (Dunn’s method). Dunn’s method comparison: NEO/BCD 4 (mg/kg) vs. NEO/BCD 16 (mg/kg); <sup>##</sup> <span class="html-italic">p</span> ≤ 0.01, and: CIM vs. NEO/BCD 16 mg/kg; <sup>###</sup> <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">Figure 5
<p>Effect of the complex NEO/βCD on mice exposed to chronic unpredictable mild stress and evaluated in the hole board test. Graphics represent (<b>A</b>) the count number, (<b>B</b>) rearings number, (<b>C</b>) head dipping time (s) and (<b>D</b>) the head dipping number. CTL, control group; FLX 2, fluoxetine at 2 mg/kg/day; NEO2, NEO/βCD at 2 mg/kg/day; NEO4, NEO/βCD at 4 mg/kg/day. Data represent the mean standard error the number of events (<b>A</b>,<b>B</b>,<b>D</b>) or seconds (<b>C</b>), of groups of 7 or 8 mice, each one. Treatment effects was analyzed with one-way analysis of variance (ANOVA), following Holm-Sidak’s method for pairwise multiple comparisons. Statistical significance is indicated as follows: ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001 versus the vehicle-control group; <sup>#</sup> <span class="html-italic">p</span> ≤ 0.05, <sup>##</sup> <span class="html-italic">p</span> ≤ 0.01 FLX vs. NEO2 and NEO4.</p>
Full article ">Figure 6
<p>Effect of NEO/βCD on the FST of animals exposed to chronic unpredictable mild stress (CUMS). (<b>A</b>) Animals administered with vehicle (VEH CUMS) and exposed to CUMS (grey bar) and animals administered with the vehicle without CUMS (VEH NO-CUMS; white bar). (<b>B</b>) All animals’ groups were exposed to CUMS and tested in the FST at the end of the treatments. CTL, control group; FLX 2, fluoxetine at 2 mg/kg/day; NEO2, NEO/βCD at 2 mg/kg/day; NEO4, NEO/βCD at 4 mg/kg/day. Data represent the mean standard error of immobility time of groups of 7 or 8 mice, each one. Treatment effect was on depressive-like behavior was analyzed with one-way analysis of variance (ANOVA) followed by the Holm-Sidak’s method for pairwise multiple comparisons, *** <span class="html-italic">p</span> ≤ 0.001 versus the vehicle-control group.</p>
Full article ">
18 pages, 8281 KiB  
Article
Study on the Mechanisms of Glrα3 in Pain Sensitization of Endometriosis
by Peiya Fan, Rong Liu, Yan Li, Shixuan Wang and Tian Li
Int. J. Mol. Sci. 2024, 25(15), 8143; https://doi.org/10.3390/ijms25158143 - 26 Jul 2024
Viewed by 537
Abstract
Endometriosis, often associated with chronic pelvic pain, can lead to anxiety and depression. This study investigates the role and mechanism of Glycine receptor alpha 3 (Glrα3) in the central sensitization of pain in endometriosis, aiming to identify new therapeutic targets. Using a Glrα3 [...] Read more.
Endometriosis, often associated with chronic pelvic pain, can lead to anxiety and depression. This study investigates the role and mechanism of Glycine receptor alpha 3 (Glrα3) in the central sensitization of pain in endometriosis, aiming to identify new therapeutic targets. Using a Glrα3 knockout mouse model of endometriosis, we employed behavioral tests, qPCR, immunofluorescence, Nissl staining, MRI, and Western blot to assess the involvement of Glrα3 in central pain sensitization. Our results indicate that endometriosis-induced hyperalgesia and anxiety–depressive-like behaviors are linked to increased Glrα3 expression. Chronic pain in endometriosis leads to gray matter changes in the sensory and insular cortices, with Glrα3 playing a significant role. The inhibition of Glrα3 alleviates pain, reduces neuronal abnormalities, and decreases glial cell activation. The absence of Glrα3 effectively regulates the central sensitization of pain in endometriosis by inhibiting glial cell activation and maintaining neuronal stability. This study offers new therapeutic avenues for the clinical treatment of endometriosis-related pain. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Behavioral test results. (<b>A</b>) Macroscopic and histological images of the ventral abdominal wall in two groups of mice (scale bar: 200 µm). H&amp;E staining of endometriotic-like lesions to confirm the success of the endometriosis model; the typical endometrioid gland, including highly cylindrical epithelium, could be seen. (<b>B</b>) Changes in thermal withdrawal latency (TWL) at weeks 4, 6, and 8 post-surgery, showing reduced TWL in the EM group compared to the Sham group. (<b>C</b>) Reductions in mechanical withdrawal threshold (MWT) in the EM group at weeks 6 and 8 post-surgery compared to the Sham group. (<b>D</b>) Representative activity tracks in the open field test at week 8 post-surgery for EM and Sham group mice. (<b>E</b>) Reduced central area activity time in the EM group compared to the Sham group in the open field test at week 8 post-surgery. (<b>F</b>) Reduced total distance traveled in the open field test by the EM group compared to the Sham group at week 4 post-surgery. (<b>G</b>) Increased immobility time in the tail suspension test for the EM group at week 8 post-surgery compared to the Sham group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. n = 5 per group.</p>
Full article ">Figure 2
<p>Differential expression of Glrα3 mRNA in brain regions. (<b>A</b>) Schematic of spatial localization in the brain regions IC, S1, HPC, and TH. (<b>B</b>) No significant difference in Glrα3 expression in the TH region between the two groups. (<b>C</b>) Increased expression of Glrα3 in the IC region of the EM group mice. (<b>D</b>) Increased expression of Glrα3 in the S1 region of the EM group mice. (<b>E</b>) No significant differences in Glrα3 expression in the HPC region between the groups. * <span class="html-italic">p</span> &lt; 0.05, ns indicates <span class="html-italic">p</span> &gt; 0.05. n = 4/5 per group.</p>
Full article ">Figure 3
<p>Behavioral test results. (<b>A</b>) No significant difference in lesion volume between EM and KO groups. (<b>B</b>) Changes in TWL detected in the hot plate test post-surgery across three groups. (<b>C</b>) Changes in TWL in the tail-flick test post-surgery across three groups. (<b>D</b>) Changes in MWT detected in the von Frey test post-surgery across three groups. (<b>E</b>) Representative activity tracks in the open field at week 12 post-surgery for three groups. (<b>F</b>) Changes in central activity time in the open field test post-surgery across three groups. (<b>G</b>) Changes in total activity distance in the open field test post-surgery across three groups. (<b>H</b>) Changes in immobility time in the tail suspension test post-surgery across three groups. Comparisons: * EM vs. KO, <sup>#</sup> EM vs. Sham, with significance levels * <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, <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, <sup>####</sup> <span class="html-italic">p</span> &lt; 0.001. n = 8 per group.</p>
Full article ">Figure 4
<p>VBM analysis differences. (<b>A</b>) Voxel comparison of gray matter volume (GMV) between EM and Sham groups. (<b>B</b>) Voxel comparison of GMV between EM and KO groups. The color bar’s top and bottom numbers indicate the t values of the statistical results. n = 8 per group.</p>
Full article ">Figure 5
<p>Differences in neuronal activity in the S1 and IC brain regions among three groups. (<b>A</b>) Representative Nissl-stained sections from the S1 and IC brain regions of three groups, scale bar 50 µm. (<b>B</b>) Nissl staining results in the S1 region. (<b>C</b>) Nissl staining results in the IC region. (<b>D</b>) Statistical results of Neun/c-Fos co-staining in the S1 region. (<b>E</b>) Statistical results of Neun/c-Fos co-staining in the IC region. (<b>F</b>) Representative sections of Neun/c-Fos co-staining in the S1 and IC brain regions, scale bar 50 µm. * <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. IOD (Integrated Optical Density). n = 4 per group.</p>
Full article ">Figure 6
<p>Activation of glial cells in the S1 and IC brain regions among three groups. (<b>A</b>) Representative Iba1-stained sections from the S1 and IC brain regions, scale bar 50 µm. (<b>B</b>) Iba1 staining results in the S1 region. (<b>C</b>) Iba1 staining results in the IC region. (<b>D</b>) GFAP staining results in the S1 region. (<b>E</b>) GFAP staining results in the IC region. (<b>F</b>) Representative GFAP-stained sections from the S1 and IC brain regions, scale bar 50 µm. * <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. n = 4 per group.</p>
Full article ">Figure 7
<p>Quantification of glial cells and inflammatory factors expression. (<b>A</b>) Protein blot analysis for Iba1 across three groups. (<b>B</b>) Protein blot analysis for GFAP across three groups. (<b>C</b>) Quantification of Iba1 mRNA levels in three groups. (<b>D</b>) Quantification of GFAP mRNA levels in three groups. (<b>E</b>) Quantification of IL1β mRNA levels in three groups. (<b>F</b>) Quantification of TNFα mRNA levels in three groups. (<b>G</b>) Quantification of IL10 mRNA levels in three groups. (<b>H</b>) Quantification of IL6 mRNA levels in three groups. * <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; 0.0001. ns: no significance. n = 4 per group.</p>
Full article ">Figure 8
<p>Schematic of the experimental design. (<b>A</b>) Schematic workflow for the first batch of experimental mice. (<b>B</b>) Schematic workflow for the second batch of experimental mice. (<b>C</b>) Detailed timeline for behavioral assessments.</p>
Full article ">
Back to TopTop