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

Journals

Article Types

Countries / Regions

Search Results (76)

Search Parameters:
Keywords = HPA axis genes

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 3446 KiB  
Article
Targeting the Kynureninase–HDAC6–Complement Axis as a Novel Therapeutic Strategy in Glioblastoma
by Arif Ul Hasan, Sachiko Sato, Mami Obara, Yukiko Kondo and Eiichi Taira
Epigenomes 2025, 9(3), 27; https://doi.org/10.3390/epigenomes9030027 - 28 Jul 2025
Viewed by 338
Abstract
Background/Objectives: Glioblastoma (GBM) is an aggressive brain tumor known for its profound heterogeneity and treatment resistance. Dysregulated complement signaling and epigenetic alterations have been implicated in GBM progression. This study identifies kynureninase (KYNU), a key enzyme in the kynurenine pathway, as a novel [...] Read more.
Background/Objectives: Glioblastoma (GBM) is an aggressive brain tumor known for its profound heterogeneity and treatment resistance. Dysregulated complement signaling and epigenetic alterations have been implicated in GBM progression. This study identifies kynureninase (KYNU), a key enzyme in the kynurenine pathway, as a novel regulator of complement components and investigates its interaction with histone deacetylase 6 (HDAC6) in the context of therapeutic targeting. Methods: KYNU expression, and its association with complement signaling in GBM, were analyzed using publicly available datasets (TCGA, GTEx, HPA). Pathway enrichment was performed via LinkedOmics. In vitro studies in GBM cell lines (U87, U251, T98G) assessed the effects of KYNU silencing and treatment with an HDAC6 inhibitor (tubastatin) and a BET inhibitor (apabetalone) on gene expression and cell viability. Results: Bioinformatic analyses revealed significant overexpression of KYNU in GBM tissues compared to normal brain tissue. KYNU expression was positively associated with genes involved in complement and coagulation cascades. In vitro experiments demonstrated that KYNU silencing reduced the expression of C3, C3AR1, and C5AR1 and suppressed GBM cell viability. Treatment with tubastatin, while reducing viability, paradoxically upregulated complement genes, suggesting potential limitations in therapeutic efficacy. However, this effect was mitigated by KYNU knockdown. Combined treatment with apabetalone and tubastatin effectively suppressed KYNU expression and enhanced cytotoxicity, particularly in cells with high complement expression. Conclusions: Our findings establish the KYNU–HDAC6–complement axis as a critical regulatory pathway in GBM. Targeting KYNU-mediated complement activation through combined epigenetic approaches—such as HDAC6 and BET inhibition—represents a promising strategy to overcome complement-driven resistance in GBM therapy. Full article
Show Figures

Figure 1

19 pages, 4255 KiB  
Article
Impacts of Early Weaning on Lamb Gut Health and Immune Function: Short-Term and Long-Term Effects
by Chong Li, Yunfei Xu, Jiale Jia, Xiuxiu Weng, Yang Zhang, Jialin Peng, Xueming An and Guoxiu Wang
Animals 2025, 15(14), 2135; https://doi.org/10.3390/ani15142135 - 18 Jul 2025
Viewed by 345
Abstract
Despite the known impacts of weaning on animal health, the underlying molecular mechanisms remain unclear, particularly how psychological and nutritional stress differentially affect gut health and immune function over time. This study hypothesized that early weaning exerts distinct short- and long-term effects on [...] Read more.
Despite the known impacts of weaning on animal health, the underlying molecular mechanisms remain unclear, particularly how psychological and nutritional stress differentially affect gut health and immune function over time. This study hypothesized that early weaning exerts distinct short- and long-term effects on lamb stress physiology, immunity, and gut health, mediated by specific molecular pathways. Twelve pairs of full-sibling male Hu sheep lambs were assigned to control (CON) or early-weaned (EW) groups. Plasma stress/immune markers were dynamically monitored, and intestinal morphology, antioxidant capacity, apoptosis, and transcriptomic profiles were analyzed at 5 and 28 days post-weaning. Early weaning triggered transient psychological stress, elevating hypothalamic–pituitary–adrenal (HPA) axis hormones (cortisol, catecholamines) and inflammatory cytokines (TNF-α) within 1 day (p < 0.05); however, stress responses were transient and recovered by 7 days post-weaning. Sustained intestinal remodeling was observed in EW lambs, featuring reduced ileal villus height, increased crypt depth (p < 0.05), and oxidative damage (MDA levels doubled vs. CON; p < 0.01). Compensatory epithelial adaptation included increased crypt depth but paradoxically reduced villus tip apoptosis. The transcriptome analysis revealed significant changes in gene expression related to immune function, fat digestion, and metabolism. Key DEGs included APOA4, linked to lipid transport adaptation; NOS2, associated with nitric oxide-mediated immune–metabolic crosstalk; and mitochondrial gene COX1, reflecting energy metabolism dysregulation. Protein–protein interaction analysis revealed NOS2 as a hub gene interacting with IDO1 and CXCL11, connecting oxidative stress to immune cell recruitment. Early weaning exerts minimal lasting psychological stress but drives persistent gut dysfunction through transcriptome-mediated changes in metabolic and immune pathways, highlighting key genes such as APOA4, NOS2, and COX1 as potential regulators of these effects. Full article
(This article belongs to the Topic Feeding Livestock for Health Improvement)
Show Figures

Figure 1

21 pages, 1089 KiB  
Review
Salivary Biomarkers as a Predictive Factor in Anxiety, Depression, and Stress
by Dana Gabriela Budala, Ionut Luchian, Dragos Ioan Virvescu, Teona Tudorici, Vlad Constantin, Zinovia Surlari, Oana Butnaru, Dan Nicolae Bosinceanu, Cosmin Bida and Monica Hancianu
Curr. Issues Mol. Biol. 2025, 47(7), 488; https://doi.org/10.3390/cimb47070488 - 26 Jun 2025
Viewed by 1012
Abstract
Anxiety and depression are highly prevalent mental health disorders often associated with dysregulation of neuroendocrine and immune systems, particularly the hypothalamic–pituitary–adrenal (HPA) axis and the sympathetic–adrenal–medullary (SAM) system. Recent research highlights the potential of salivary biomarkers to serve as non-invasive indicators for psychological [...] Read more.
Anxiety and depression are highly prevalent mental health disorders often associated with dysregulation of neuroendocrine and immune systems, particularly the hypothalamic–pituitary–adrenal (HPA) axis and the sympathetic–adrenal–medullary (SAM) system. Recent research highlights the potential of salivary biomarkers to serve as non-invasive indicators for psychological distress. This narrative review synthesizes current evidence on key salivary biomarkers, cortisol, alpha-amylase (sAA), secretory immunoglobulin A (sIgA), chromogranin A (CgA), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), C-reactive protein (CRP), brain-derived neurotrophic factor (BDNF), and salivary microRNAs (miRNAs), in relation to anxiety, depression, and stress. A comprehensive literature search (2010–2025) was conducted using multiple databases and relevant MeSH terms. The review reveals consistent associations between these salivary analytes and stress-related disorders, reflecting changes in neuroendocrine activity, immune response, and neuroplasticity. Cortisol and sAA mirror acute stress reactivity, while cytokines and CRP indicate chronic inflammation. BDNF and miRNAs provide insight into neuroplastic dysfunction and gene regulation. Despite promising results, limitations such as variability in sampling methods and biomarker specificity remain. In conclusion, salivary biomarkers offer a promising avenue for early detection, monitoring, and personalization of treatment in mood and anxiety disorders. Conclusions: Cortisol and alpha-amylase serve as the principal markers of acute stress response, whereas cytokines such as IL-6 and TNF-α, together with CRP, indicate chronic inflammation associated with extended emotional distress. Full article
Show Figures

Figure 1

23 pages, 17920 KiB  
Article
Comparative Analysis of HPA-Axis Dysregulation and Dynamic Molecular Mechanisms in Acute Versus Chronic Social Defeat Stress
by Jiajun Yang, Yifei Jia, Ting Guo, Siqi Zhang, Jin Huang, Huiling Lu, Leyi Li, Jiahao Xu, Gefei Liu and Ke Xiao
Int. J. Mol. Sci. 2025, 26(13), 6063; https://doi.org/10.3390/ijms26136063 - 24 Jun 2025
Viewed by 555
Abstract
Organisms respond to environmental stress primarily through the autonomic nervous system and hypothalamic–pituitary–adrenal (HPA) axis, regulating metabolism, psychological states, and immune function and modulating memory, reward processing, and immune responses. The HPA axis plays a central role in stress response, exhibiting distinct activation [...] Read more.
Organisms respond to environmental stress primarily through the autonomic nervous system and hypothalamic–pituitary–adrenal (HPA) axis, regulating metabolism, psychological states, and immune function and modulating memory, reward processing, and immune responses. The HPA axis plays a central role in stress response, exhibiting distinct activation patterns under acute versus chronic social defeat stress. However, differences in physiological impacts and regulatory pathways between these stress conditions remain understudied. This study integrates RNA sequencing and behavioral analyses to reveal that acute social defeat stress triggers transient anxiety-like behaviors, accompanied by systemic inflammation and immediate-early gene (IEG) activation. In contrast, chronic social defeat stress induces long-term behavioral and physiological alterations, including neurotransmitter imbalance (e.g., reduced GABA and increased glutamate), sustained activation of maladaptive pathways (e.g., IL-17 signaling), and disrupted corticosterone synthesis. These findings highlight the dynamic regulatory role of the HPA axis under varying stress conditions, providing novel insights into mental health disorders such as anxiety and depression. The study identifies potential therapeutic targets to mitigate chronic social defeat stress effects and offers a theoretical foundation for personalized interventions. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

37 pages, 17348 KiB  
Article
Anxiolytic and Antidepressant Effects of Organic Polysulfide, Dimethyl Trisulfide Are Partly Mediated by the Transient Receptor Potential Ankyrin 1 Ion Channel in Mice
by Kitti Göntér, Viktória Kormos, Erika Pintér and Gábor Pozsgai
Pharmaceutics 2025, 17(6), 781; https://doi.org/10.3390/pharmaceutics17060781 - 14 Jun 2025
Viewed by 775
Abstract
Background/Objectives: Dimethyl trisulfide (DMTS) is a naturally occurring polysulfide with known antioxidant and neuroprotective properties. DMTS is a lipophilic transient receptor potential ankyrin 1 (TRPA1) ligand that reaches the central nervous system (CNS). Its role in the CNS, particularly regarding depression-like behaviour, [...] Read more.
Background/Objectives: Dimethyl trisulfide (DMTS) is a naturally occurring polysulfide with known antioxidant and neuroprotective properties. DMTS is a lipophilic transient receptor potential ankyrin 1 (TRPA1) ligand that reaches the central nervous system (CNS). Its role in the CNS, particularly regarding depression-like behaviour, has yet to be explored. This study investigates the influence of DMTS on stress responses and whether this effect is mediated through the TRPA1 ion channel, known for its role in stress adaptation. Using a mouse model involving three-week exposure, we examined the impact of DMTS on depression-like behaviour and anxiety and identified the involved brain regions. Methods: Our methods involved testing both Trpa1-wild-type and gene-knockout mice under CUMS conditions and DMTS treatment. DMTS was administered intraperitoneally at a dose of 30 mg/kg on days 16 and 20 of the 21-day CUMS protocol—in hourly injections seven times to ensure sustained exposure. Various behavioural assessments—including the open field, marble burying, tail suspension, forced swim, and sucrose preference tests—were performed to evaluate anxiety and depression-like behaviour. Additionally, we measured body weight changes and the relative weights of the thymus and adrenal glands, while serum levels of corticosterone and adrenocorticotropic hormone were quantified via ELISA. FOSB (FBJ murine osteosarcoma viral oncogene homolog B) immunohistochemistry was utilised to assess chronic neuronal activation in stress-relevant brain areas. Results: Results showed that CUMS induces depression-like behaviour, with the response being modulated by the TRPA1 status and that DMTS treatment significantly reduced these effects when TRPA1 channels were functional. DMTS also mitigated thymus involution due to hypothalamic–pituitary–adrenal (HPA) axis dysregulation. Conclusions: Overall, DMTS appears to relieve depressive and anxiety symptoms through TRPA1-mediated pathways, suggesting its potential as a dietary supplement or adjunct therapy for depression and anxiety. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Graphical abstract

18 pages, 1224 KiB  
Article
Reduced Gene Dosage of the Psychiatric Risk Gene Cacna1c Is Associated with Impairments in Hypothalamic–Pituitary–Adrenal Axis Activity in Rats
by Anna L. Moon, Eleanor R. Mawson, Patricia Gasalla, Lawrence S. Wilkinson, Dominic M. Dwyer, Jeremy Hall and Kerrie L. Thomas
Int. J. Mol. Sci. 2025, 26(12), 5547; https://doi.org/10.3390/ijms26125547 - 10 Jun 2025
Viewed by 459
Abstract
Common and rare variation in CACNA1C gene expression has been consistently associated with neuropsychiatric disorders such as schizophrenia, bipolar disorder, and major depression. However, the underlying biological pathways that cause this association have yet to be fully determined. In this study, we present [...] Read more.
Common and rare variation in CACNA1C gene expression has been consistently associated with neuropsychiatric disorders such as schizophrenia, bipolar disorder, and major depression. However, the underlying biological pathways that cause this association have yet to be fully determined. In this study, we present evidence that rats with a reduced gene dosage of Cacna1c have increased basal corticosterone levels in the periphery and reduced the expression of Nr3c1 encoding the glucocorticoid receptor in the hippocampus and hypothalamus. These results are consistent, with an effect of Cacna1c dosage on hypothalamus–pituitary–adrenal (HPA) axis function. Heterozygous Cacna1c rats had lower levels of the histone markers H3K4me3 and H3K27acat exon 17 of the Nr3c1 gene. These histone modifications are typically linked to increased gene expression, but here were not associated with changes in the expression of exon 17 variants under non-stress conditions. Heterozygous Cacna1c rats additionally show increased anxiety behaviours. These results support an association of Cacna1c heterozygosity with the altered activity of the HPA axis and function in the resting state, and this may be a predisposing mechanism that contributes to the increased risk of psychiatric disorders with stress. Full article
Show Figures

Figure 1

26 pages, 433 KiB  
Review
Hyperarousal, Dissociation, Emotion Dysregulation and Re-Experiencing—Towards Understanding Molecular Aspects of PTSD Symptoms
by Aleksandra Brzozowska and Jakub Grabowski
Int. J. Mol. Sci. 2025, 26(11), 5216; https://doi.org/10.3390/ijms26115216 - 29 May 2025
Viewed by 1369
Abstract
Approximately 70% of people will experience a traumatic event in their lifetime, but post-traumatic stress disorder (PTSD) will only develop in 3.9% and complex post-traumatic stress disorder (CPTSD) in 1–8% of the population worldwide, although in some countries (e.g., Poland and Northern Ireland) [...] Read more.
Approximately 70% of people will experience a traumatic event in their lifetime, but post-traumatic stress disorder (PTSD) will only develop in 3.9% and complex post-traumatic stress disorder (CPTSD) in 1–8% of the population worldwide, although in some countries (e.g., Poland and Northern Ireland) it will develop in a much higher percentage. Stress-related disorders have a complex pathogenesis involving neurophysiological, genetic, epigenetic, neuroendocrine and environmental factors. This article reviews the current state of knowledge on the molecular aspects of selected PTSD symptoms: hypervigilance, re-experiencing, emotion dysregulation and dissociation, i.e., the symptoms with strong neurobiological components. Among analysed susceptibility factors are specific gene polymorphisms (e.g., FKBP5, COMT, CHRNA5, CRHR1, 5-HTTLPR, ADCY8 and DRD2) and their interactions with the environment, changes in the HPA axis, adrenergic hyperactivity and disturbances in the activity of selected anatomical structures (including the amygdala, prefrontal cortex, corpus callosum, anterior cingulate gyrus and hippocampus). It is worth noting that therapeutic methods with proven effectiveness in PTSD (TF-CBT and EMDR) have a substantial neurobiological rationale. Molecular aspects seem crucial when searching for effective screening/diagnostic methods and new potential therapeutic options. Full article
21 pages, 4078 KiB  
Article
The Effects and Mechanisms of Continuous 7-Day Hypobaric Hypoxia Exposure on Sleep Architecture in Rats
by Fang Li, Xianxie Zhang, Anping Ye, Ling Qi, Tianke Huang, Xitai Chen, Maoxing Li, Chengrong Xiao, Yuguang Wang, Yue Gao and Zengchun Ma
Int. J. Mol. Sci. 2025, 26(11), 4998; https://doi.org/10.3390/ijms26114998 - 22 May 2025
Viewed by 680
Abstract
High-altitude environments pose significant risks for insomnia development, which severely compromises both physiological health and occupational performance. To elucidate the mechanisms underlying altitude-induced sleep disruption and establish a validated animal model for therapeutic intervention development, we exposed Sprague-Dawley rats to hypobaric hypoxia (5500 [...] Read more.
High-altitude environments pose significant risks for insomnia development, which severely compromises both physiological health and occupational performance. To elucidate the mechanisms underlying altitude-induced sleep disruption and establish a validated animal model for therapeutic intervention development, we exposed Sprague-Dawley rats to hypobaric hypoxia (5500 m altitude equivalent: 308 mmHg, 20.37% O2, PiO2 8.0 kPa) for 7 days. We employed continuous wireless telemetry to monitor EEG/EMG signals, with concurrent analysis of physiological parameters, blood biochemistry, histopathology, transcriptomics, and protein expression. Quantitative analyses demonstrated decreased caloric intake, transient body mass reduction, and immune-metabolic disturbances. While total sleep duration showed no significant variation, sleep architecture displayed elevated wakefulness periods, reduced active wakefulness, a decreasing trend of slow-wave sleep (SWS), and increased paradoxical sleep (PS) accompanied by attenuated circadian oscillations. The duration of SWS episodes was significantly shortened, indicating a sleep homeostasis imbalance that peaked on day 3. Biochemical profiling revealed reduced levels of antioxidant enzymes, elevated pro-inflammatory cytokines, and hypothalamic–pituitary–adrenal axis activation. Transcriptomic analyses identified the critical involvement of serotonergic/glutamatergic synaptic regulation, lipid metabolism, IL-17 signaling, and cortisol synthesis pathways. Western blot analyses confirmed OX2R upregulation, 5-HT1AR downregulation, and circadian gene dysregulation. Our findings demonstrate that hypobaric hypoxia induces sleep disruption via coordinated mechanisms involving oxidative stress, inflammatory activation, HPA axis hyperactivity, neurotransmitter imbalance, and circadian clock dysfunction, providing a robust preclinical model for mechanistic exploration and therapeutic target identification. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

25 pages, 2404 KiB  
Article
Network Pharmacology-Guided Evaluation of Ginger and Cornelian Cherry Extracts Against Depression and Metabolic Dysfunction in Estrogen-Deficient Chronic Stressed Rats
by Nara Lee, Ting Zhang, Hanbin Joe and Sunmin Park
Int. J. Mol. Sci. 2025, 26(10), 4829; https://doi.org/10.3390/ijms26104829 - 18 May 2025
Viewed by 893
Abstract
This study investigated the therapeutic effects of water extracts from Zingiber officinale Roscoe (ginger) and Cornus officinalis Siebold and Zucc. fruits (COF) water extracts on depression-like behavior and metabolic dysfunction in estrogen-deficient rats exposed to chronic mild stress (CMS). Network pharmacology analysis identified [...] Read more.
This study investigated the therapeutic effects of water extracts from Zingiber officinale Roscoe (ginger) and Cornus officinalis Siebold and Zucc. fruits (COF) water extracts on depression-like behavior and metabolic dysfunction in estrogen-deficient rats exposed to chronic mild stress (CMS). Network pharmacology analysis identified three bioactive compounds in ginger and four in COF, with 11 overlapping targets linked to both depression and metabolic pathways, primarily involving NR3C1, HTR2A, MAOA, and SLC6A4 genes associated with hypothalamic–pituitary–adrenal (HPA) axis regulation and neurotransmitter modulation. Ovariectomized rats received 200 mg/kg/day of ginger or COF extracts for 7 weeks, with a 4-week CMS protocol initiated at week 3. Both extracts significantly improved depression-like behaviors, memory performance, glucose tolerance, lipid profiles, and bone mineral density, normalized HPA axis markers (corticosterone and ACTH), and increased hippocampal serotonin and dopamine levels. Ginger demonstrated greater efficacy in improving memory and metabolic outcomes compared to COF. Molecular docking further validated these findings, revealing strong and stable interactions between key phytochemicals—such as hydroxygenkwanin and telocinobufagin—and target proteins MAOA, HTR2A, and NR3C1, supporting their mechanistic role in stress and mood regulation. These results support supplementing ginger and COF extracts as promising botanical interventions for estrogen-deficiency-related mood and metabolic disorders, with potential clinical application at a human-equivalent dose of 1.5 g/day. Full article
(This article belongs to the Special Issue Medicinal Plants and Bioactive Compounds in Health and Disease)
Show Figures

Figure 1

20 pages, 6264 KiB  
Article
Profile of Rat Adrenal microRNAs Induced by Gonadectomy and Testosterone or Estradiol Replacement
by Karol Jopek, Marianna Tyczewska, Małgorzata Blatkiewicz, Anna Olechnowicz, Marta Szyszka, Ewelina Stelcer, Sylwia Ciesiółka, Maria Jopek, Ludwik K. Malendowicz and Marcin Ruciński
Int. J. Mol. Sci. 2025, 26(10), 4543; https://doi.org/10.3390/ijms26104543 - 9 May 2025
Viewed by 538
Abstract
Sex-related differences in the structure and function of the adrenal cortex in mature rats are well recognized, largely driven by the action of sex hormones on the hypothalamic–pituitary–adrenal axis (HPA). By replacing testosterone or estradiol in gonadectomized rats, we aimed to elucidate the [...] Read more.
Sex-related differences in the structure and function of the adrenal cortex in mature rats are well recognized, largely driven by the action of sex hormones on the hypothalamic–pituitary–adrenal axis (HPA). By replacing testosterone or estradiol in gonadectomized rats, we aimed to elucidate the regulation of micro RNA (miRNA) profiles by sex hormones and their role in physiological adrenal function, providing new insights into gene expression modulation in the adrenal gland. This paper focuses on the description of miRNA profiles using the microarray technique. In our study, we observed significant sex differences in miRNA and mRNA expression levels. These differences are as follows: miRNA expression profiles Male C vs. Female C-0 down, 25 up-regulated, while mRNA profiles were 43 down and 27 up-regulated. Moreover, we observed the most significant differences in miRNA profiles between orchiectomized male rats supplemented with testosterone (ORX + T) and ovariectomized female rats treated with estradiol (OVX + E). Furthermore, we described changes in target gene expression and biological processes regulated by miRNAs. The processes most differentially expressed between the ORX + T and OVX + E groups are those related to the metabolism and synthesis of sterol compounds, the positive and negative regulation of metabolic processes in cells, e.g., cholesterol metabolism, response to various external factors, e.g., hormones, regulation of processes related to cell motility. We also identified several miRNAs, such as miR-370, miR-377, and miR-503, that exhibited interesting changes in their expression after testosterone or estradiol replacement. These results contribute to a deeper understanding of adrenal physiology. Full article
Show Figures

Figure 1

21 pages, 830 KiB  
Review
Bridging ADHD and Metabolic Disorders: Insights into Shared Mechanisms and Clinical Implications
by Ilaria Marcelli, Umberto Capece and Alfredo Caturano
Diabetology 2025, 6(5), 40; https://doi.org/10.3390/diabetology6050040 - 8 May 2025
Viewed by 4924
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterized by inattention, impulsivity and/or hyperactivity. In recent years, metabolic alterations, primarily obesity, insulin resistance, and diabetes, have emerged as frequent comorbidities in individuals with ADHD, suggesting a bidirectional relationship between neurodevelopmental and metabolic dysfunctions. Emerging [...] Read more.
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterized by inattention, impulsivity and/or hyperactivity. In recent years, metabolic alterations, primarily obesity, insulin resistance, and diabetes, have emerged as frequent comorbidities in individuals with ADHD, suggesting a bidirectional relationship between neurodevelopmental and metabolic dysfunctions. Emerging evidence indicates that dysregulation of dopaminergic signaling, disturbances in the hypothalamic-pituitary-adrenal (HPA) axis, and chronic low-grade inflammation are central to both ADHD symptomatology and metabolic impairments. For instance, alterations in dopamine-related genes (e.g., DRD4, DAT1) not only affect cognitive and behavioral functions but also play a role in appetite regulation and glucose homeostasis. Epidemiological studies further demonstrate that individuals with ADHD exhibit poorer glycemic control and a higher prevalence of both type 1 and type 2 diabetes, while early-life metabolic challenges such as maternal diabetes may predispose offspring to ADHD. This review aims to comprehensively synthesize the epidemiological, genetic, and pathogenetic evidence linking ADHD to metabolic alterations. We discuss key pathophysiological pathways—including dopaminergic dysregulation, HPA axis disturbances, inflammation, and oxidative stress—and evaluate their contributions to the co-occurrence of ADHD and metabolic disorders. In addition, we explore the clinical implications and integrated treatment approaches that encompass lifestyle modifications, pharmacological therapies, and multidisciplinary care. Finally, we outline future research directions to develop personalized and holistic interventions. Full article
Show Figures

Figure 1

25 pages, 4786 KiB  
Article
Exploring the Toxicity and Therapeutic Potential of A. dahurica and A. pubescens in Zebrafish Larvae: Insights into Anxiety Treatment Mechanisms
by Mariola Herbet, Jarosław Widelski, Marta Ostrowska-Leśko, Anna Serefko, Krzysztof Wojtanowski, Joanna Kurek and Iwona Piątkowska-Chmiel
Int. J. Mol. Sci. 2025, 26(7), 2884; https://doi.org/10.3390/ijms26072884 - 22 Mar 2025
Viewed by 609
Abstract
This study assessed the toxicity and therapeutic potential of Angelica dahurica and Angelica pubescens using Danio rerio (zebrafish) larvae. Toxicity was evaluated through mortality, malformations, and gene expression changes related to stress and the HPA axis. A. dahurica demonstrated low toxicity (LD50 (50% [...] Read more.
This study assessed the toxicity and therapeutic potential of Angelica dahurica and Angelica pubescens using Danio rerio (zebrafish) larvae. Toxicity was evaluated through mortality, malformations, and gene expression changes related to stress and the HPA axis. A. dahurica demonstrated low toxicity (LD50 (50% lethal dose) >200 µg/mL), with no significant malformations at 15–30 µg/mL, although higher doses caused edemas and heart defects. A. pubescens exhibited higher toxicity, with 100% mortality at 200 µg/mL and severe malformations. Both species showed potential cardiotoxicity, slowing heart rates after prolonged exposure. Gene expression studies suggested A. dahurica had stress-protective effects, increasing nr3c1 expression, while A. pubescens had dose-dependent effects, with lower concentrations having anxiolytic properties and higher concentrations increasing stress. Interestingly, diazepam showed unexpected gene expression changes, highlighting the influence of environmental and dosage factors. In conclusion, both species show therapeutic potential for anxiety, with A. dahurica showing promising effects at lower concentrations. However, A. pubescens requires careful dosage management due to its higher toxicity risks. Further studies are needed to optimize therapeutic applications and fully understand mechanisms of action. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

85 pages, 24685 KiB  
Review
Adaptogens in Long-Lasting Brain Fatigue: An Insight from Systems Biology and Network Pharmacology
by Alexander Panossian, Terrence Lemerond and Thomas Efferth
Pharmaceuticals 2025, 18(2), 261; https://doi.org/10.3390/ph18020261 - 15 Feb 2025
Cited by 1 | Viewed by 8416
Abstract
Long-lasting brain fatigue is a consequence of stroke or traumatic brain injury associated with emotional, psychological, and physical overload, distress in hypertension, atherosclerosis, viral infection, and aging-related chronic low-grade inflammatory disorders. The pathogenesis of brain fatigue is linked to disrupted neurotransmission, the glutamate-glutamine [...] Read more.
Long-lasting brain fatigue is a consequence of stroke or traumatic brain injury associated with emotional, psychological, and physical overload, distress in hypertension, atherosclerosis, viral infection, and aging-related chronic low-grade inflammatory disorders. The pathogenesis of brain fatigue is linked to disrupted neurotransmission, the glutamate-glutamine cycle imbalance, glucose metabolism, and ATP energy supply, which are associated with multiple molecular targets and signaling pathways in neuroendocrine-immune and blood circulation systems. Regeneration of damaged brain tissue is a long-lasting multistage process, including spontaneously regulating hypothalamus-pituitary (HPA) axis-controlled anabolic–catabolic homeostasis to recover harmonized sympathoadrenal system (SAS)-mediated function, brain energy supply, and deregulated gene expression in rehabilitation. The driving mechanism of spontaneous recovery and regeneration of brain tissue is a cross-talk of mediators of neuronal, microglia, immunocompetent, and endothelial cells collectively involved in neurogenesis and angiogenesis, which plant adaptogens can target. Adaptogens are small molecules of plant origin that increase the adaptability of cells and organisms to stress by interaction with the HPA axis and SAS of the stress system (neuroendocrine-immune and cardiovascular complex), targeting multiple mediators of adaptive GPCR signaling pathways. Two major groups of adaptogens comprise (i) phenolic phenethyl and phenylpropanoid derivatives and (ii) tetracyclic and pentacyclic glycosides, whose chemical structure can be distinguished as related correspondingly to (i) monoamine neurotransmitters of SAS (epinephrine, norepinephrine, and dopamine) and (ii) steroid hormones (cortisol, testosterone, and estradiol). In this narrative review, we discuss (i) the multitarget mechanism of integrated pharmacological activity of botanical adaptogens in stress overload, ischemic stroke, and long-lasting brain fatigue; (ii) the time-dependent dual response of physiological regulatory systems to adaptogens to support homeostasis in chronic stress and overload; and (iii) the dual dose-dependent reversal (hormetic) effect of botanical adaptogens. This narrative review shows that the adaptogenic concept cannot be reduced and rectified to the various effects of adaptogens on selected molecular targets or specific modes of action without estimating their interactions within the networks of mediators of the neuroendocrine-immune complex that, in turn, regulates other pharmacological systems (cardiovascular, gastrointestinal, reproductive systems) due to numerous intra- and extracellular communications and feedback regulations. These interactions result in polyvalent action and the pleiotropic pharmacological activity of adaptogens, which is essential for characterizing adaptogens as distinct types of botanicals. They trigger the defense adaptive stress response that leads to the extension of the limits of resilience to overload, inducing brain fatigue and mental disorders. For the first time, this review justifies the neurogenesis potential of adaptogens, particularly the botanical hybrid preparation (BHP) of Arctic Root and Ashwagandha, providing a rationale for potential use in individuals experiencing long-lasting brain fatigue. The review provided insight into future research on the network pharmacology of adaptogens in preventing and rehabilitating long-lasting brain fatigue following stroke, trauma, and viral infections. Full article
(This article belongs to the Section Natural Products)
Show Figures

Graphical abstract

40 pages, 4205 KiB  
Article
Evaluation of Prenatal Transportation Stress on DNA Methylation (DNAm) and Gene Expression in the Hypothalamic–Pituitary–Adrenal (HPA) Axis Tissues of Mature Brahman Cows
by Audrey L. Earnhardt-San, Emilie C. Baker, Kubra Z. Cilkiz, Rodolfo C. Cardoso, Noushin Ghaffari, Charles R. Long, Penny K. Riggs, Ronald D. Randel, David G. Riley and Thomas H. Welsh
Genes 2025, 16(2), 191; https://doi.org/10.3390/genes16020191 - 4 Feb 2025
Cited by 1 | Viewed by 922
Abstract
Background/Objectives: The experience of prenatal stress results in various physiological disorders due to an alteration of an offspring’s methylome and transcriptome. The objective of this study was to determine whether PNS affects DNA methylation (DNAm) and gene expression in the stress axis tissues [...] Read more.
Background/Objectives: The experience of prenatal stress results in various physiological disorders due to an alteration of an offspring’s methylome and transcriptome. The objective of this study was to determine whether PNS affects DNA methylation (DNAm) and gene expression in the stress axis tissues of mature Brahman cows. Methods: Samples were collected from the paraventricular nucleus (PVN), anterior pituitary (PIT), and adrenal cortex (AC) of 5-year-old Brahman cows that were prenatally exposed to either transportation stress (PNS, n = 6) or were not transported (Control, n = 8). The isolated DNA and RNA samples were, respectively, used for methylation and RNA-Seq analyses. A gene ontology and KEGG pathway enrichment analysis of each data set within each sample tissue was conducted with the DAVID Functional Annotation Tool. Results: The DNAm analysis revealed 3, 64, and 99 hypomethylated and 2, 93, and 90 hypermethylated CpG sites (FDR < 0.15) within the PVN, PIT, and AC, respectively. The RNA-Seq analysis revealed 6, 25, and 5 differentially expressed genes (FDR < 0.15) in the PVN, PIT, and AC, respectively, that were up-regulated in the PNS group relative to the Control group, as well as 24 genes in the PIT that were down-regulated. Based on the enrichment analysis, several developmental and cellular processes, such as maintenance of the actin cytoskeleton, cell motility, signal transduction, neurodevelopment, and synaptic function, were potentially modulated. Conclusions: The methylome and transcriptome were altered in the stress axis tissues of mature cows that had been exposed to prenatal transportation stress. These findings are relevant to understanding how prenatal experiences may affect postnatal neurological functions. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

27 pages, 29104 KiB  
Article
Metabonomics and Transcriptomics Analyses Reveal the Underlying HPA-Axis-Related Mechanisms of Lethality in Larimichthys polyactis Exposed to Underwater Noise Pollution
by Qinghua Jiang, Yu Zhang, Ting Ye, Xiao Liang and Bao Lou
Int. J. Mol. Sci. 2024, 25(23), 12610; https://doi.org/10.3390/ijms252312610 - 24 Nov 2024
Viewed by 1060
Abstract
The problem of marine noise pollution has a long history. Strong noise (>120 dB re 1 µPa) will affects the growth, development, physiological responses, and behaviors of fish, and also can induce the stress response, posing a mortal threat. Although many studies have [...] Read more.
The problem of marine noise pollution has a long history. Strong noise (>120 dB re 1 µPa) will affects the growth, development, physiological responses, and behaviors of fish, and also can induce the stress response, posing a mortal threat. Although many studies have reported that underwater noise may affect the survival of fish by disturbing their nervous system and endocrine system, the underlying causes of death due to noise stimulation remain unknown. Therefore, in this study, we used the underwater noise stress models to conduct underwater strong noise (50–125 dB re 1 µPa, 10–22,000 Hz) stress experiments on small yellow croaker for 10 min (short-term noise stress) and 6 days (long-term noise stress). A total of 150 fishes (body weight: 40–60 g; body length: 12–14 cm) were used in this study. Omics (metabolomics and transcriptomics) studies and quantitative analyses of important genes (HPA (hypothalamic–pituitary–adrenal)-axis functional genes) were performed to reveal genetic and metabolic changes in the important tissues associated with the HPA axis (brain, heart, and adrenal gland). Finally, we found that the strong noise pollution can significantly interfere with the expression of HPA-axis functional genes (including corticotropin releasing hormone (CRH), corticotropin releasing hormone receptor 2 (CRHR2), and arginine vasotocin (AVT)), and long-term stimulation can further induce metabolic disorders of the functional tissues (brain, heart, and adrenal gland), posing a lethal threat. Meanwhile, we also found that there were two kinds of death processes, direct death and chronic death, and both were closely related to the duration of stimulation and the regulation of the HPA axis. Full article
(This article belongs to the Special Issue Fish Nutrition, Metabolism and Physiology)
Show Figures

Figure 1

Back to TopTop