1
|
Su C, Huang T, Zhang M, Zhang Y, Zeng Y, Chen X. Glucocorticoid receptor signaling in the brain and its involvement in cognitive function. Neural Regen Res 2025; 20:2520-2537. [PMID: 39248182 PMCID: PMC11801288 DOI: 10.4103/nrr.nrr-d-24-00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-adrenal axis regulates the secretion of glucocorticoids in response to environmental challenges. In the brain, a nuclear receptor transcription factor, the glucocorticoid receptor, is an important component of the hypothalamic-pituitary-adrenal axis's negative feedback loop and plays a key role in regulating cognitive equilibrium and neuroplasticity. The glucocorticoid receptor influences cognitive processes, including glutamate neurotransmission, calcium signaling, and the activation of brain-derived neurotrophic factor-mediated pathways, through a combination of genomic and non-genomic mechanisms. Protein interactions within the central nervous system can alter the expression and activity of the glucocorticoid receptor, thereby affecting the hypothalamic-pituitary-adrenal axis and stress-related cognitive functions. An appropriate level of glucocorticoid receptor expression can improve cognitive function, while excessive glucocorticoid receptors or long-term exposure to glucocorticoids may lead to cognitive impairment. Patients with cognitive impairment-associated diseases, such as Alzheimer's disease, aging, depression, Parkinson's disease, Huntington's disease, stroke, and addiction, often present with dysregulation of the hypothalamic-pituitary-adrenal axis and glucocorticoid receptor expression. This review provides a comprehensive overview of the functions of the glucocorticoid receptor in the hypothalamic-pituitary-adrenal axis and cognitive activities. It emphasizes that appropriate glucocorticoid receptor signaling facilitates learning and memory, while its dysregulation can lead to cognitive impairment. This provides clues about how glucocorticoid receptor signaling can be targeted to overcome cognitive disability-related disorders.
Collapse
Affiliation(s)
- Chonglin Su
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Taiqi Huang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Meiyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Xingxing Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Mancini GF, Blasi E, Marchetta E, Morena M, Borgi M, Campolongo P. The impact of stress on fear memory retention: A meta-analysis of rodent fear conditioning studies. Neurosci Biobehav Rev 2025; 175:106221. [PMID: 40409442 DOI: 10.1016/j.neubiorev.2025.106221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 05/17/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Pavlovian fear conditioning is a widely used behavioural task for studying fear memory in rodents. During conditioning, rodents learn to associate a conditioned stimulus (e.g., context or tone; contextual or auditory fear conditioning, CFC or AFC, respectively) with an aversive one (e.g., footshock), resulting in a conditioned fear response. Fear memory retention is assessed thorough freezing behaviour, a species-specific defensive reaction, observed during exposure to the conditioned stimulus alone. Fear memory is influenced by sex and stress, with stress exposure prior to conditioning potentially inducing maladaptive fear responses. This meta-analysis examines how pre-conditioning stress exposure modulates memory retention in rodents. Across N = 94 studies included, we analyzed freezing behaviour based on several factors: type of paradigm (CFC vs AFC), species (rat vs mouse), sex (male vs female), stress type (physical vs pharmacological vs psychological vs combination of two or more stressors type), stress duration (acute or chronic), stress timing (prenatal vs early postnatal vs adolescence vs adulthood). The results indicate that stress significantly enhances contextual conditioned freezing behaviour. Stress-induced effects in CFC models vary across species but are not sex-specific. Additionally, these effects are influenced by stress-related factors. These findings highlight the importance of considering multiple variables when studying stress and fear memory processes, offering valuable insights for improving clinical approaches to fear memory-related diseases (e.g., post-traumatic stress disorder).
Collapse
Affiliation(s)
- Giulia Federica Mancini
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| | - Eleonora Blasi
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| | - Enrico Marchetta
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| | - Maria Morena
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy; Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, Rome 00143, Italy
| | - Marta Borgi
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Patrizia Campolongo
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy; Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, Rome 00143, Italy.
| |
Collapse
|
3
|
van Swieten M, de Looff P, VanDerNagel J, Bouwmeester S, Didden R. Listening to music is associated with reduced physiological and subjective stress in people with mild intellectual disabilities: A biofeedback study. RESEARCH IN DEVELOPMENTAL DISABILITIES 2025; 161:104976. [PMID: 40138868 DOI: 10.1016/j.ridd.2025.104976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Many people with mild intellectual disabilities are at increased risk to experience stress. Reducing stress is important because experiencing prolonged and elevated stress can have detrimental effects on mental and physical health and it is associated with aggressive behaviour and self-harm. AIMS This preliminary study investigated whether an intervention combining biofeedback with listening to music is effective and whether a personalized music playlist is more effective than self-selected music in reducing physiological and subjective stress in participants with mild intellectual disabilities. METHODS We collected 103 music listening sessions over a period of 2-4 weeks for 11 participants throughout their daily routines. They listened to music when they received biofeedback on their increased stress level (as measured by wearable biosensor Nowatch) or when they themselves felt stressed. Participants listened either to self-selected music or to a personalised playlist compiled with X-system, music technology that predicts the effect of a song on levels of autonomic arousal. Pulse rate (PR) and skin conductance level (SCL) were measured with the EmbracePlus and subjective feelings of stress and mood were measured with two scale questions. After the intervention phase, participants and their caregivers completed a short questionnaire to evaluate their experiences with using the X-system playlist. RESULTS Mixed regression analyses showed reductions in PR and SCL during listening to music, and indications were found for reductions in subjective stress and improvement of mood after intervention. Listening to music compiled with X-system was not more effective than listening to self-selected music. However, lower combined arousal values (a feature of X-system) from self-selected and X-system music predicted lower PR and SCL, indicating that these indices can be used to select songs that have a relaxing or energizing effect. CONCLUSIONS AND IMPLICATIONS The present study suggests that music listening is associated with both subjective and physiological stress reduction. Listening to music might be an accessible, inexpensive and empowering strategy for stress reduction and improving emotion regulation, which could also benefit mental and physical health. Several challenges were encountered while implementing the intervention and suggestions for future research are given.
Collapse
Affiliation(s)
- Marlieke van Swieten
- Behavioural Science Institute, Radboud University, Nijmegen 6525 GD, the Netherlands; Research & Development, Trajectum, Zwolle 8025 AV, the Netherlands.
| | - Peter de Looff
- Behavioural Science Institute, Radboud University, Nijmegen 6525 GD, the Netherlands; Science and Treatment Innovation, Fivoor, Rotterdam 3014 AE, the Netherlands; National Expert Centre Intellectual Disabilities and Severe Behavioral Problems, De Borg, Bilthoven 3723 MB, the Netherlands; Department of Developmental Psychology, Tilburg University, Tilburg 5037 AB, the Netherlands
| | - Joanneke VanDerNagel
- Tactus, Deventer 7400 AD, the Netherlands; Aveleijn, Borne 7622 GW, the Netherlands; Department Human Media Interactions, University of Twente, Enschede 7500 AE, the Netherlands; Nijmegen Institute for Scientist-Practitioners in Addiction, Radboud University, Nijmegen 6503 GK, the Netherlands
| | - Samantha Bouwmeester
- Department of Developmental Psychology, Tilburg University, Tilburg 5037 AB, the Netherlands; Out of the BoxPlot, Rotterdam, The Netherlands
| | - Robert Didden
- Behavioural Science Institute, Radboud University, Nijmegen 6525 GD, the Netherlands; Research & Development, Trajectum, Zwolle 8025 AV, the Netherlands
| |
Collapse
|
4
|
Zhang L, Tuoliken H, Li J, Gao H. Diet, gut microbiota, and health: a review. Food Sci Biotechnol 2025; 34:2087-2099. [PMID: 40351733 PMCID: PMC12064509 DOI: 10.1007/s10068-024-01759-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/19/2024] [Accepted: 11/13/2024] [Indexed: 05/14/2025] Open
Abstract
The relationship between diet and human physical and mental health is highly interconnected and has been significantly correlated with the occurrence of various diseases, including neurological disorders, cancer, and chronic inflammatory diseases. Moreover, diet has been demonstrated to play a pivotal role in governing gut microbiota composition, making it one of the most influential factors. The diet is crucial in connecting humans and their gut microorganisms. The nutrients ingested supply energy to the body and serve as substrates for the metabolic processes of the gut microorganisms. Consequently, the gut flora and their metabolites reciprocally impact the host's metabolism, thereby influencing the physiological state of the human body. Extensive investigations on human and mouse models have revealed that diet potentially underlies various effects on human health and disease. Graphical abstract
Collapse
Affiliation(s)
- Longxiang Zhang
- The Second Department of Gastroenterology, the First Affiliated Hospital of Xinjiang, Medical University, Urumqi, 830000 Xinjiang China
| | - Haishaer Tuoliken
- The Second Department of Gastroenterology, the First Affiliated Hospital of Xinjiang, Medical University, Urumqi, 830000 Xinjiang China
| | - Jian Li
- The Second Department of Gastroenterology, the First Affiliated Hospital of Xinjiang, Medical University, Urumqi, 830000 Xinjiang China
| | - Hongliang Gao
- The Second Department of Gastroenterology, the First Affiliated Hospital of Xinjiang, Medical University, Urumqi, 830000 Xinjiang China
| |
Collapse
|
5
|
Tsuda S, Hou J, Thompson FJ, Bose PK. Traumatic brain injury-induced anxiety: Injury and plasticity of the central noradrenergic system. Exp Neurol 2025; 388:115182. [PMID: 39929384 DOI: 10.1016/j.expneurol.2025.115182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 03/17/2025]
Abstract
Long-term anxiety is a hallmark symptom following traumatic brain injury (TBI). Although the central noradrenergic system (CNAS) is known to play a critical role in anxiety by regulating the excitability of several intricately interconnected brain structures via its projections to them, critical questions remain regarding the nature and extent of TBI-induced neuroplastic alterations in the CNAS and how these alterations relate to anxiety disorders. Knowledge relative to these questions is pivotal to development and refinement of therapies for TBI-associated anxiety disorders, including post-traumatic stress disorder. To this end, this study was designed to determine the impacts of chronic TBI on neuroplasticity of the CNAS and their significance in anxiety disorders in a clinically relevant rodent model. A standardized weight-drop model was used to produce controlled impacts of mild-to-moderate TBI in rats. Following the elevated plus maze tests to longitudinally assess anxiety-like behavior at 2 and 18 weeks post-injury of TBI animals, brain tissues of naïve and TBI rats were coronally sectioned and immunostained for a noradrenergic (NA) marker (dopamine β-hydroxylase) and neuronal nuclei in the central NA production sites and critical anxiety-regulating brain structures. We discovered that TBI caused robust losses of NA cells in the locus coeruleus and NA innervation of the central nucleus of the amygdala, an emotional processing center. Conversely, TBI caused intense gains of NA cells in the A2/A1 cell groups and NA innervation of other major anxiety-regulating regions. These changes coincided with progressively elevated anxiety-like behavior. Possibly, NA properties of A2/A1 cells were upregulated to compensate for the TBI-induced severe cell losses in the locus coeruleus. We conclude that these bi-directional vast alterations in the CNAS following chronic TBI contribute to dysregulated anxiety and, in part, the pathophysiology of human post-traumatic stress disorder.
Collapse
Affiliation(s)
- Shigeharu Tsuda
- Department of Anesthesiology, University of Florida, 1600 SW Archer Rd m509, Gainesville, FL 32610, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville, FL 32608, USA
| | - Jiamei Hou
- Department of Anesthesiology, University of Florida, 1600 SW Archer Rd m509, Gainesville, FL 32610, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville, FL 32608, USA
| | - Floyd J Thompson
- Department of Neuroscience, University of Florida, 1149 Newell Dr, Gainesville, FL 32610, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville, FL 32608, USA
| | - Prodip K Bose
- Department of Anesthesiology, University of Florida, 1600 SW Archer Rd m509, Gainesville, FL 32610, USA; Department of Neurology, University of Florida, 1149 Newell Dr, Gainesville, FL 32611, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville, FL 32608, USA.
| |
Collapse
|
6
|
Topuz RD, Cevık B, Guler K, Gunduz O, Karadag CH, Ulugol A. Do serum endocannabinoid and N-acylethanolamine concentrations reflect their brain levels in two different rat stress models? World J Biol Psychiatry 2025; 26:199-210. [PMID: 40366731 DOI: 10.1080/15622975.2025.2502394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Although the brain is not the only source of circulating endocannabinoids and their levels can be affected by many factors, it is underlined that serum endocannabinoid levels can be used as a biomarker in psychiatric disorders. In this study, we aimed to examine whether serum endocannabinoid and N-acylethanolamine concentrations reflect their brain levels. METHODS In the present study acute immobilisation (AIS) and post-traumatic stress (PTSD) models were applied to Wistar albino male rats. Rota rod performance, forced swim, open field and elevated plus maze tests were performed. Endocannabinoid and N-acylethanolamine levels in serum and hippocampus, amygdala and cortex were assessed using LC-MS/MS. RESULTS We observed significant increases in anandamide (AEA), palmitoylethanolamide (PEA) and oleoethylethanolamide (OEA) levels in the amygdala and hippocampus in both models except PEA in amygdala in the AIS group, while 2-AG levels decreased. There was no change in serum AEA and 2-AG levels in all groups; in the PTSD group serum PEA levels were higher whereas OEA levels were lower in both the AIS and the PTSD groups. CONCLUSION Our results show that there is no correlation in endocannabinoid and N-acylethanolamine levels between serum and specific brain regions in two stress models of rat.
Collapse
Affiliation(s)
- Ruhan Deniz Topuz
- Faculty of Medicine, Department of Medical Pharmacology, Trakya University, Edirne, Turkey
| | - Buse Cevık
- Faculty of Medicine, Department of Medical Pharmacology, Trakya University, Edirne, Turkey
| | - Kursat Guler
- Faculty of Medicine, Department of Medical Pharmacology, Trakya University, Edirne, Turkey
| | - Ozgur Gunduz
- Faculty of Medicine, Department of Medical Pharmacology, Trakya University, Edirne, Turkey
| | - Cetin Hakan Karadag
- Faculty of Medicine, Department of Medical Pharmacology, Trakya University, Edirne, Turkey
| | - Ahmet Ulugol
- Faculty of Medicine, Department of Medical Pharmacology, Trakya University, Edirne, Turkey
| |
Collapse
|
7
|
Padilla VJ, Scheffrahn K, Muñiz V, Lorenz TK, Elkins G. Heart Rate Variability, Hypnosis, and Psychotherapy. Appl Psychophysiol Biofeedback 2025; 50:289-295. [PMID: 39625562 DOI: 10.1007/s10484-024-09679-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 05/16/2025]
Abstract
Hypnotherapy is increasingly recognized as an effective treatment option for a wide range of psychological conditions and the psychological elements of physiologic health concerns. Despite its growing use, the mechanisms underlying hypnotherapy's therapeutic benefits remain unknown. Heart rate variability (HRV), a non-invasive biomarker of autonomic nervous system (ANS) activity and other processes relevant to the psychophysiological stress response, has been used to study the physiological effects of hypnosis. HRV is associated with adaptability to stress and overall mental and physical health. We review how HRV changes during hypnosis treatment and propose mechanisms by which hypnotherapy influences the stress response and psychological flexibility and improves wellbeing. Additionally, we highlight the use of HRV biofeedback as an adjunct to hypnotherapy and psychotherapy and illustrate its potential to enhance therapeutic outcomes. To showcase practical applications, we present a case study of hypnotherapy's impact on HRV in the treatment of hot flashes in post-menopausal women. We conclude by calling for further empirical research, particularly randomized controlled trials, to solidify the integration of HRV metrics in hypnotherapy practice. Understanding the interaction between hypnotherapy and HRV will support more targeted and effective interventions, benefiting both clinicians and patients.
Collapse
Affiliation(s)
- Victor Julian Padilla
- Department of Psychology and Neuroscience, Baylor University, 801 Washington Ave, 2nd Floor, Waco, TX, USA
| | - Katherine Scheffrahn
- Department of Psychology and Neuroscience, Baylor University, 801 Washington Ave, 2nd Floor, Waco, TX, USA
| | - Vanessa Muñiz
- Department of Psychology and Neuroscience, Baylor University, 801 Washington Ave, 2nd Floor, Waco, TX, USA
| | - Tierney K Lorenz
- Department of Psychology, Center for Brain, Biology and Behavior, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Gary Elkins
- Department of Psychology and Neuroscience, Baylor University, 801 Washington Ave, 2nd Floor, Waco, TX, USA.
| |
Collapse
|
8
|
Birnie MT, Baram TZ. The evolving neurobiology of early-life stress. Neuron 2025; 113:1474-1490. [PMID: 40101719 PMCID: PMC12097948 DOI: 10.1016/j.neuron.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 03/20/2025]
Abstract
Because early-life stress is common and constitutes a strong risk factor for cognitive and mental health disorders, it has been the focus of a multitude of studies in humans and experimental models. Yet, we have an incomplete understanding of what is perceived as stressful by the developing brain, what aspects of stress influence brain maturation, what developmental ages are particularly vulnerable to stress, which molecules mediate the effects of stress on brain operations, and how transient stressful experiences can lead to enduring emotional and cognitive dysfunctions. Here, we discuss these themes, highlight the challenges and progress in resolving them, and propose new concepts and avenues for future research.
Collapse
Affiliation(s)
- Matthew T Birnie
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA; Department of Neurology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
9
|
Hu Y, Li Z, Zhu Y, Xing M, Xie X, Zhao P, Cheng X, Xiao C, Xia Y, Wu J, Luo Y, Ko H, Tang Y, Ye X, Lin WJ. Microglial repopulation reverses radiation-induced cognitive dysfunction by restoring medial prefrontal cortex activity and modulating leukotriene-C4 synthesis. Acta Neuropathol Commun 2025; 13:105. [PMID: 40390112 PMCID: PMC12087111 DOI: 10.1186/s40478-025-02026-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Cranial radiotherapy and environmental radiation exposure are associated with increased risk of cognitive dysfunction, including memory deficits and mood disorders, yet the underlying mechanisms remain poorly understood. In this study, we demonstrate that cranial irradiation induces hypoactivity in the medial prefrontal cortex (mPFC) of mice, leading to anxiety-like behaviors and memory impairments, which can be prevented by optogenetic activation of mPFC excitatory neurons. Radiaiton exposure also causes a significant reduction in microglial density within the mPFC, accompanied by morphological and transcriptional alterations in the remaining microglia. Notably, microglial repopulation, achieved through CSF1R antagonist-mediated depletion prior to irradiation and subsequent repopulation, restores mPFC neuronal acitivity and reverses cognitive and behavioral deficits. Integrated bulk RNA sequencing and microglial proteomic analysis of the mPFC reveal that microglial repopulation specifically modulates the leukotriene-C4 biosynthesis pathway, without significant changes in canonical pro-inflammatory cytokines or chemokines. Importantly, pharmacological inhibition of leukotriene-C4 synthase ameliorates radiation-induced anxiety and memory impairments. These findings identify leukotriene-C4 signaling as a critical mechanism underlying radiation-induced cognitive dysfunction and suggest that microglial repopulation and targted inhibition of leukotriene-C4 represent potential therapeutic strategies for mitigating radiation-associated cognitive disorders.
Collapse
Affiliation(s)
- Yubo Hu
- Medical College of Jiaying University, Meizhou, Guangdong, 514031, China
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhe Li
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yafeng Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Mengdan Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China
| | - Xiaoru Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China
| | - Panwu Zhao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xin Cheng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Chuan Xiao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yuting Xia
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jingru Wu
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yuan Luo
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Gerald Choa Neuroscience Center, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Chow Yuk Ho Technology Center for Innovative Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
- Peter Hung Pain Research Institute, Faculty of Medicine, The Chinese University of Hong Kong, SAR, Hong Kong, China
| | - Yamei Tang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Wei-Jye Lin
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
10
|
Meier M, Dimitroff SJ, Denk BF, Unternaehrer E, Pruessner JC. Effect of sweet and caloric drinks on cardiac reactivity to slow-paced breathing in healthy adults. Sci Rep 2025; 15:17368. [PMID: 40389457 PMCID: PMC12089329 DOI: 10.1038/s41598-025-00980-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 05/02/2025] [Indexed: 05/21/2025] Open
Abstract
Cardiac reactivity is proposed to be a central indicator of autonomic functioning. While hyperglycemia plays a limited role in cardiac stress reactivity, it is unclear whether it may modulate cardiac reactivity in non-stressful situations. We investigated the effect of glucose on cardiac reactivity to a relaxing exercise, namely, slow-paced breathing (SPB). A total of 115 adults (age mean = 23.28 years, SD = 6.88; 76% female) either consumed a sweet & caloric, a sweet, a caloric drink, or pure water after baseline. Later, they performed a sustained attention test and SPB. Electrocardiography and impedance cardiography was obtained, and blood glucose and subjective relaxation were measured repeatedly. We analyzed changes in parasympathetic (root mean square of successive differences [RMSSD]) and sympathetic (pre-ejection period [PEP]) cardiac activity and subjective relaxation using growth curve models and performed correlational analyses. Hyperglycemia triggered cardiac PNS withdrawal and SNS activation. Despite this, SPB increased cardiac PNS activity and subjective relaxation and decreased cardiac SNS activity in all groups. Our results align with the autonomic space model and highlight the tight link between autonomic regulation and blood glucose homeostasis. Hyperglycemia might play a limited modulating role in cardiac reactivity to slow-paced breathing.
Collapse
Affiliation(s)
| | | | - Bernadette F Denk
- University of Konstanz, Constance, Germany
- Centre for the Advanced Study of Collective Behavior, Constance, Germany
| | - Eva Unternaehrer
- Child- and Adolescent Psychiatric Research Department, University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
| | - Jens C Pruessner
- University of Konstanz, Constance, Germany
- Centre for the Advanced Study of Collective Behavior, Constance, Germany
| |
Collapse
|
11
|
Liu F, He Z, Wang Y. Neural mechanisms, influencing factors and interventions in empathic pain. Neuropharmacology 2025; 269:110349. [PMID: 39914620 DOI: 10.1016/j.neuropharm.2025.110349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/10/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Empathic pain, defined as the emotional resonance with the suffering of others, is akin to the observer's own experience of pain and is vital for building and sustaining positive interpersonal relationships. Despite its importance, the neural mechanism of empathic pain remains poorly understood. In this review, we integrated and summarized the currently knowledge on the neural networks associated with empathic pain, focusing on key brain regions such as the insula, anterior cingulate cortex (ACC), ventral tegmental area (VTA), nucleus accumbens (NAc), and locus coeruleus (LC)/norepinephrine (NE)-sympatho-adrenomedullar (LC/NE-SAM) system. We also reviewed the factors that affect empathic pain, including gender, personal beliefs, the intimacy of relationships, and the nature of interpersonal relationships, and highlighted the central role of the insula and ACC in the neural circuitry of empathy, the importance of the IC-BLA and ACC-NAc/VTA connections in modulating empathic pain, and the involvement of the LC/NE-SAM system in mediating pain empathy. We further discussed how gender significantly influences empathic pain, with women showing more intense emotional reactions to social distress than men. It also summarized the roles of personal pain history and empathy levels in modulating empathic responses. Furthermore, the review emphasized the impact of social factors such as the nature of interpersonal relationships and experiences of social exclusion on empathic pain. By providing a detailed exploration of the neural mechanisms and influencing factors of empathic pain, this review aims to establish a robust foundation for developing targeted therapeutic strategies and improving pain management in clinical settings.
Collapse
Affiliation(s)
- Furui Liu
- School of Pharmacy, Hangzhou Normal University, 311121, Zhejiang, China
| | - Ziwan He
- School of Pharmacy, Hangzhou Normal University, 311121, Zhejiang, China
| | - Yongjie Wang
- School of Pharmacy, Hangzhou Normal University, 311121, Zhejiang, China.
| |
Collapse
|
12
|
Moreland M, Curry C, Wang A, Vansickel M, Wu Z, Sieberg CB. Somatosensory and prefrontal cortex activity relates to emotional outcomes and hair cortisol concentration in chronic postsurgical pain. Sci Rep 2025; 15:16304. [PMID: 40348822 PMCID: PMC12065869 DOI: 10.1038/s41598-025-00685-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/26/2025] [Indexed: 05/14/2025] Open
Abstract
Chronic post-surgical pain (CPSP) poses significant socioeconomic and humanitarian challenges. This study investigated relationships between resting-state neural activation in the somatosensory cortex (SMC) and emotional functioning outcomes (depression, anxiety, perceived stress), and between prefrontal cortex (PFC) activation and chronic stress, measured by hair cortisol concentration (HCC); and whether pain intensity moderates these relationships in females with CPSP. Twenty-nine females with CPSP reported baseline pain, completed emotional functioning questionnaires, underwent functional Near-Infrared Spectroscopy, and provided hair samples for HCC analysis. Pearson's correlation examined associations between emotional functioning and SMC activation, and between HCC and PFC activation. Benjamini-Hochberg correction adjusted for multiple comparisons. Significant correlations were further tested using moderation analyses to assess whether pain intensity influenced these associations. Left SMC activation was positively correlated with depressive symptoms (r = 0.505, pFDR = 0.036) and anxiety(r = 0.705, pFDR = 0.039). Right lateral PFC activation showed a negative correlation with HCC (r = -0.475, pFDR = 0.048). Pain intensity did not significantly moderate these relationships. Findings suggest associations between brain activity and emotional functioning in females with CPSP, highlighting potential neural targets for future interventions. This study supports the utility of multimodal approaches to further phenotype CPSP and inform precision medicine strategies.
Collapse
Affiliation(s)
- Margaret Moreland
- Department of Psychiatry, Center for Health Outcomes and Interdisciplinary Research, Massachusetts General Hospital, One Bowdoin Square, Boston, MA, 02114, USA
| | - Caitlin Curry
- Department of Psychiatry, Center for Health Outcomes and Interdisciplinary Research, Massachusetts General Hospital, One Bowdoin Square, Boston, MA, 02114, USA
| | - Anthony Wang
- Department of Psychiatry, Center for Health Outcomes and Interdisciplinary Research, Massachusetts General Hospital, One Bowdoin Square, Boston, MA, 02114, USA
| | - Madison Vansickel
- Department of Psychiatry, Center for Health Outcomes and Interdisciplinary Research, Massachusetts General Hospital, One Bowdoin Square, Boston, MA, 02114, USA
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ziyan Wu
- Department of Psychiatry, Center for Health Outcomes and Interdisciplinary Research, Massachusetts General Hospital, One Bowdoin Square, Boston, MA, 02114, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Christine B Sieberg
- Department of Psychiatry, Center for Health Outcomes and Interdisciplinary Research, Massachusetts General Hospital, One Bowdoin Square, Boston, MA, 02114, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Division of Adolescent and Young Adult Medicine, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
13
|
Garces-Arilla S, Hidalgo V, Fidalgo C, Salvador A, Juan MC, Mendez-Lopez M. Post-encoding stress and spatial memory consolidation: No significant associations with cortisol and DHEA reactivity. Behav Brain Res 2025; 485:115525. [PMID: 40049334 DOI: 10.1016/j.bbr.2025.115525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025]
Abstract
Memory consolidation is enhanced by post-encoding stress via cortisol, although the role of dehydroepiandrosterone (DHEA) remains uncertain. This study investigated the effect of the Maastricht Acute Stress Test (MAST) on psychological and hormonal (salivary cortisol and DHEA) responses and performance on a virtual reality object-location memory (OLM) task. The association between hormonal reactivity and OLM task performance was also investigated. Fifty-four participants aged 18-23 were randomly assigned to a stress group (n = 30) and a control group (n = 24). Participants completed an encoding trial of the OLM task and the MAST/control procedure in an acquisition session. A retrieval session 24 h later included verbal object and visual location recognition tests, as well as an object-context binding (OCB) trial. Results showed that the stress group reported significantly higher state anxiety and negative affect after the MAST, perceived as more stressful, painful, and unpleasant. This group also exhibited a significant increase in cortisol and DHEA levels in response to the MAST. Controlling for age, participants in the stress condition made fewer errors in recognising semantically unrelated objects and tended to complete the OCB trial in a shorter time. In addition, no associations were found between cortisol or DHEA reactivity and memory performance in either the stress group or the control group. Our data indicate that post-encoding stress enhances consolidation; however, no association was found between hormonal reactivity and this process, suggesting that these endocrine responses do not directly support the observed improvement in memory consolidation.
Collapse
Affiliation(s)
- Sara Garces-Arilla
- Department of Psychology and Sociology, University of Zaragoza, Faculty of Social and Human Sciences, Teruel, Spain.
| | - Vanesa Hidalgo
- Department of Psychology and Sociology, University of Zaragoza, Faculty of Social and Human Sciences, Teruel, Spain; Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, Valencia, Spain; Instituto de Investigación Sanitaria Aragón (IIS), Zaragoza, Aragon, Spain.
| | - Camino Fidalgo
- Department of Psychology and Sociology, University of Zaragoza, Faculty of Social and Human Sciences, Teruel, Spain; Instituto de Investigación Sanitaria Aragón (IIS), Zaragoza, Aragon, Spain.
| | - Alicia Salvador
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, Valencia, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain.
| | - M-Carmen Juan
- Instituto Universitario de Automática e Informática Industrial, Universitat Politècnica de València, Valencia, Spain.
| | - Magdalena Mendez-Lopez
- Department of Psychology and Sociology, University of Zaragoza, Faculty of Social and Human Sciences, Teruel, Spain; Instituto de Investigación Sanitaria Aragón (IIS), Zaragoza, Aragon, Spain.
| |
Collapse
|
14
|
Chen C, Xiong B, Tan W, Tian Y, Zhang S, Wu J, Song P, Qin S. Setting the tone for the day: Cortisol awakening response proactively modulates fronto-limbic circuitry for emotion processing. Neuroimage 2025; 315:121251. [PMID: 40345506 DOI: 10.1016/j.neuroimage.2025.121251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 04/04/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025] Open
Abstract
The cortisol awakening response (CAR) has been linked to a variety of emotion-related psychiatric conditions and is proposed to prepare the brain for upcoming stress and challenges. Yet, the underlying neurobiological mechanisms of such proactive effects on emotional processing remain elusive. In the current double-blinded, pharmacologically-manipulated study, 36 male adults (DXM group) received cortisol-repressive dexamethasone on the previous night, then performed the Emotional Face Matching Task (EFMT) during fMRI scanning the next afternoon. Relative to the placebo group (31 male adults), the DXM group exhibited lower accuracy in the emotion matching condition, but not in the sensorimotor control condition. Psychophysiological interaction (PPI) analyses revealed significant task-by-group interaction involving the right and left amygdala, but not the medial orbitofrontal cortex (MOFC) or hippocampus. Specifically, the DXM group exhibited stronger functional connectivity between the right amygdala and left dorsolateral prefrontal cortex (lDLPFC) during emotion condition but reduced connectivity in the same network during control condition, as compared to the placebo group. Meanwhile, the DXM group exhibited weaker left amygdala-right posterior middle temporal gyrus (rMTG) connectivity than the placebo group during control condition, but there was no group effect in the connectivity during emotion condition. These results indicate that the CAR proactively modulates fronto-limbic functional organization for emotion processing in male adults. Our findings support a causal link between CAR and its proactive effects on emotional processing, and suggest a model of CAR-mediated brain preparedness where CAR sets a tonic tone for the upcoming day to actively regulate neuroendocrinological responses to emotionally charged stimuli on a moment-to-moment basis.
Collapse
Affiliation(s)
- Changming Chen
- School of Educational Sciences, Chongqing Normal University, Chongqing, 401331, China
| | - Bingsen Xiong
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Faculty of Psychology, Beijing Normal University, Beijing 100875, China
| | - Wenlong Tan
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Faculty of Psychology, Beijing Normal University, Beijing 100875, China
| | - Yanqiu Tian
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Faculty of Psychology, Beijing Normal University, Beijing 100875, China
| | - Shouwen Zhang
- Neuroelectrophysiology Department, Beijing DaWangLu Emergency Hospital, Beijing, 100122, China.
| | - Jianhui Wu
- Shenzhen Key Laboratory of Affective and Social Cognitive Science, Shenzhen University, Shenzhen, 518060, China
| | - Peng Song
- Department of Medical Oncology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Shaozheng Qin
- Beijing Key Laboratory of Applied Experimental Psychology, National Demonstration Center for Experimental Psychology Education, Faculty of Psychology, Beijing Normal University, Beijing 100875, China; State Key Laboratory of Cognitive Neuroscience and Learning & International Data Group/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; Chinese Institute for Brain Research, Beijing, 100069, China.
| |
Collapse
|
15
|
Olchanyi MD, Schreier DR, Li J, Maffei C, Sorby-Adams A, Kinney HC, Healy BC, Freeman HJ, Shless J, Destrieux C, Tregidgo H, Iglesias JE, Brown EN, Edlow BL. Probabilistic Mapping and Automated Segmentation of Human Brainstem White Matter Bundles. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.01.25326687. [PMID: 40385397 PMCID: PMC12083584 DOI: 10.1101/2025.05.01.25326687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Brainstem white matter bundles are essential conduits for neural signaling involved in modulation of vital functions ranging from homeostasis to human consciousness. Their architecture forms the anatomic basis for brainstem connectomics, subcortical mesoscale circuit models, and deep brain navigation tools. However, their small size and complex morphology compared to cerebral white matter structures makes mapping and segmentation challenging in neuroimaging. This results in a near absence of automated brainstem white matter tracing methods. We leverage diffusion MRI tractography to create BrainStem Bundle Tool (BSBT), which segments eight key white matter bundles in the rostral brainstem. BSBT performs automated segmentation on a custom probabilistic fiber map generated from tractography with a convolutional neural network architecture tailored for detection of small structures. We demonstrate BSBTs robustness across diffusion MRI acquisition protocols through validation on healthy subject in vivo scans and ex vivo scans of brain specimens with corresponding histology. Using BSBT, we reveal distinct brainstem white matter bundle alterations in Alzheimer's disease, Parkinson's disease, and acute traumatic brain injury cohorts through tract-based analysis and classification tasks. Finally, we provide proof-of-principle evidence supporting the prognostic utility of BSBT in a longitudinal analysis of coma recovery. BSBT creates opportunities to automatically map brainstem white matter in large imaging cohorts and investigate its role in a broad spectrum of neurological disorders.
Collapse
Affiliation(s)
- Mark D. Olchanyi
- Neuroscience Statistics Research Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David R. Schreier
- Neuroscience Statistics Research Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jian Li
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Chiara Maffei
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | | | - Hannah C. Kinney
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian C. Healy
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- T.H Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Holly J. Freeman
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Jared Shless
- Neuroscience Statistics Research Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christophe Destrieux
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France
- CHRU de Tours, 2 Boulevard Tonnellé, Tours, France
| | | | - Juan Eugenio Iglesias
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Hawkes Institute, University College London, London, UK
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emery N. Brown
- Neuroscience Statistics Research Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA
| | - Brian L. Edlow
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
16
|
Raucourt L, Masson S. The Effect of Gabapentin on the Efficiency of a Desensitization-Counter-Conditioning Claw-Trimming Protocol for Cats with Healthcare Phobias: A Double-Blind, Placebo-Controlled Crossover Trial. Animals (Basel) 2025; 15:1326. [PMID: 40362141 PMCID: PMC12070960 DOI: 10.3390/ani15091326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Feline healthcare phobia is a major challenge in veterinary practice, limiting medical care and increasing stress for cats, owners, and veterinarians. Traditional desensitization-counter-conditioning (DS-CC) protocols aim to reduce fear responses, but their success is limited in sensitized cats. This study evaluates whether gabapentin can improve the efficiency of DS-CC training in healthcare phobia in cats. Forty-two sensitized cats participated in a claw-trimming learning protocol. Using a double-blind, placebo-controlled crossover trial, the cats followed 10 training sessions: the first 5 under one product and the last 5 under the other one. Their progression through the learning steps was monitored. The results indicate that gabapentin significantly accelerated the cats' ability to complete DS-CC steps, suggesting a positive effect on the learning process. While some side effects, such as mild sedation and ataxia, were observed in 42% of the cats, they were transient and did not hinder participation. These findings suggest that gabapentin should be added when engaging patients in DS-CC protocols in order to lower the time needed to achieve therapy, which will improve the overall welfare of our patients. Further studies are needed to confirm these results in other types of therapies and in a clinical setting.
Collapse
Affiliation(s)
- Lua Raucourt
- Oniris—École Nationale Vétérinaire, Agroalimentaire et de L’alimentation, 44300 Nantes, France;
| | - Sylvia Masson
- Clinique Vétérinaire de la Tivollière, 38565 Voreppe, France
- No Ledge Research, 38340 Voreppe, France
| |
Collapse
|
17
|
Maková M, Kašparová S, Bačiak L, Gogola D, Sumbalová Z, Brucknerová I, Bukatová S, Dubovický M. Effects of maternal depression and antidepressant treatment on neurotransmitters, brain regions, and mitochondrial function in rat dams. Neurochem Int 2025; 187:105981. [PMID: 40319913 DOI: 10.1016/j.neuint.2025.105981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Increasing evidence suggests that mothers experience stress before or during pregnancy, which can significantly impact their GABAergic system and lead to amygdala hyperactivity. While animal models are expected to reflect the above findings in humans, the current knowledge on the effects of chronic unpredictable mild stress (CUMS) in rat dams remains insufficient. Therefore, the objective of this study was to investigate the structural and neurochemical alterations in the dorsal hippocampus, specifically gamma-aminobutyric acid (GABA) and glutamate (Glu) relative to total creatine (tCr), induced by the CUMS and the effects of antidepressant mirtazapine (MIR) treatment. Magnetic resonance imaging and proton localized magnetic resonance spectroscopy were used in rat dams at two time points to assess the reversibility of these alterations. Eight weeks post-CUMS, chronic stress induced significant alterations in hippocampal metabolism and structural changes, including lower GABA/tCr concentrations and an increased amygdala volume compared to controls. In stressed dams treated with MIR, no changes in GABA levels or amygdala volume were observed. Fourteen weeks post-CUMS, no significant changes in hippocampal neurochemistry were confirmed, while amygdala changes persisted in stressed dams. Moreover, significant time-dependent changes were observed in the amygdala and hypothalamus in the control group with MIR. Interestingly, high-resolution respirometry was performed to assess brain mitochondrial function, revealing only changes in this group. Based on these findings, we confirmed the reversibility of metabolite. Furthermore, MIR has demonstrated potential in regulating neurotransmitter levels and protecting amygdala volume after stress; however, further research is needed to fully understand its therapeutic effects.
Collapse
Affiliation(s)
- Marianna Maková
- Institute of Physical Chemistry and Chemical Physics, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Slovak Republic; Central Laboratories, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Slovak Republic
| | - Svatava Kašparová
- Central Laboratories, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Slovak Republic.
| | - Ladislav Bačiak
- Central Laboratories, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Slovak Republic
| | - Daniel Gogola
- Institute of Measurement Science, Slovak Academy Sciences, Slovak Republic
| | - Zuzana Sumbalová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University in Bratislava, Slovak Republic
| | - Ingrid Brucknerová
- Neonatal Department of Intensive Medicine in Bratislava, Faculty of Medicine, Comenius University in Bratislava, Slovak Republic; National Institute of Children's Diseases, Bratislava, Slovak Republic
| | - Stanislava Bukatová
- Centre of Experimental Medicine of the Slovak Academy of Sciences, Institute of Experimental Pharmacology and Toxicology, Bratislava, Slovak Republic
| | - Michal Dubovický
- Centre of Experimental Medicine of the Slovak Academy of Sciences, Institute of Experimental Pharmacology and Toxicology, Bratislava, Slovak Republic
| |
Collapse
|
18
|
Xiang YT, Ma J, Wu JJ, Xue X, Gao X, Hua XY, Zheng MX, Xu JG. Brain-thyroid crosstalk: 18F-FDG-PET/MRI evidence in patients with follicular thyroid adenomas. Brain Res Bull 2025; 224:111324. [PMID: 40157550 DOI: 10.1016/j.brainresbull.2025.111324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/15/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVE The hypothalamic-pituitary-thyroid axis has been well-known. However, whether follicular thyroid adenoma (FTA) could affect brain glucose metabolism is still unknown. Therefore, we explored the brain glucose metabolic characteristics of FTA with Fluorodeoxyglucose F18 positron emission tomography/magnetic resonance imaging. METHODS Totally 30 FTA patients without clinical symptoms (FTA group), and 60 age- and sex-matched healthy controls (HC group) were included and randomly divided into cohort A and B in 2:1 ratio. Cohort A was analyzed with scaled sub-profile model/principal component analysis (SSM/PCA) for pattern identification. Cohort B was calculated the individual scores to validate expression of the pattern. Then we calculated the metabolic connectivity based on characteristics of the pattern to investigate the underlying mechanism. Finally, we constructed metabolic brain networks and analyzed the topological properties to further explore the brain metabolic model. RESULTS In SSM/PCA, FTA group showed an almost global, left-right symmetrical pattern. In metabolic connectivity, FTA group showed increased metabolic connectivity in brain regions of the sensorimotor network, ventral default mode network (DMN), posterior salient network, right executive control network (ECN), visuospatial network and language network when compared to HC group, and showed decreased connectivity in dorsal DMN and left ECN. In topological properties of brain network, FTA group showed an increased betweenness centrality (BC) in left rolandic operculum, a decreased BC in superior temporal gyrus, increased BC and Degree in right precentral gyrus, increased D in right parahippocampal gyrus and left hippocampus, and decreased D and efficiency in right orbital part of middle frontal gyrus (FDR correction for multiple comparisons, P < 0.05). CONCLUSION Although FTA patients are not yet symptomatic, their brain metabolic characteristics include extensive brain alterations, disrupted internal connectivity, not only involving brain regions associated with endocrine activity, but also brain networks and regions associated with motor, emotion and cognition.
Collapse
Affiliation(s)
- Yun-Ting Xiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, China
| | - Jie Ma
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, China; Department of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia-Jia Wu
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, China; Department of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Xue
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, China; Department of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Gao
- Universal Medical Imaging Diagnostic Center, Shanghai, China
| | - Xu-Yun Hua
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, China; Department of Traumatology and Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Mou-Xiong Zheng
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, China; Department of Traumatology and Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jian-Guang Xu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, China; Department of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
19
|
Cardaci V, Carminati M, Tondello M, Pecorino B, Serretti A, Zanardi R. Understanding and treating postpartum depression: a narrative review. Int Clin Psychopharmacol 2025; 40:127-137. [PMID: 38941162 DOI: 10.1097/yic.0000000000000560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Postpartum depression (PPD) is an increasingly prevalent but still poorly characterized disorder. Causal and modulating factors include hormones fluctuations, such as estrogen, progesterone, and allopregnolone, pathways imbalances, such as oxytocin and kynurenine, chronobiological factors, and brain imaging alterations. Treatment may differ from the traditional major depression management, while selective serotonin reuptake inhibitors such as sertraline are commonly used and suggested by guidelines, neurosteroids such as brexanolone and the more convenient zuranolone have been recently approved. Newer neurosteroids such as ganaxolone, valaxanolone, and lysaxanolone are currently under development, but also esketamine and psychedelics are promising potential treatments. Other somatic treatments including brain stimulation techniques and light therapy also showed benefit. PPD is therefore increasingly understood as, at least partially, independent from major depressive disorder. Specific and individualized treatments including pharmacological and non-pharmacological therapies are progressively being introduced in the routine clinical practice.
Collapse
Affiliation(s)
- Vincenzo Cardaci
- Department of Clinical Neurosciences, Vita-Salute San Raffaele University, Milan
| | - Matteo Carminati
- Department of Clinical Neurosciences, Vita-Salute San Raffaele University, Milan
| | - Mattia Tondello
- Department of Clinical Neurosciences, Vita-Salute San Raffaele University, Milan
| | - Basilio Pecorino
- Department of Medicine and Surgery, Kore University of Enna, Enna
| | | | - Raffaella Zanardi
- Department of Clinical Neurosciences, Vita-Salute San Raffaele University, Milan
- Department of Psychiatry, Mood Disorder Unit, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
20
|
Hanchate NK. Single-cell genomics meets systems neuroscience: Insights from mapping the brain circuitry of stress. J Neuroendocrinol 2025; 37:e70005. [PMID: 39956535 PMCID: PMC12045673 DOI: 10.1111/jne.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 12/26/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
Responses to external and internal dangers is essential for survival and homeostatic regulation. Hypothalamic corticotropin-releasing hormone neurons (CRHNs) play a pivotal role in regulating neuroendocrine responses to fear and stress. In recent years, the application of neurogenetic tools, such as fiber photometry, chemogenetics and optogenetics, have provided new insights into the dynamic neuronal responses of CRHNs during stressful events, offering new perspectives into their functional significance in mediating neurobehavioural responses to stress. Transsynaptic viral tracers have facilitated the comprehensive mapping of neuronal inputs to CRHNs. Furthermore, the development and application of innovative single-cell genomic tools combined with viral tracing have begun to pave the way for a deeper understanding of the transcriptional profiles of neural circuit components, enabling molecular-anatomical circuit mapping. Here, I will discuss how these systems neuroscience approaches and novel single-cell genomic methods are advancing the molecular and functional mapping of stress neurocircuits, their associated challenges and future directions.
Collapse
Affiliation(s)
- Naresh K. Hanchate
- Genetics & Genomic Medicine DepartmentUCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
| |
Collapse
|
21
|
Smail MA, Cotella EM, Martelle SE, Chambers JB, Parikh RK, Moore CE, Packard BA, Nawreen N, Moloney RD, Herman JP. Regulation of behavioral responses to single prolonged stress in male and female rats: Role of PACAP. Neurobiol Stress 2025; 36:100727. [PMID: 40524691 PMCID: PMC12168213 DOI: 10.1016/j.ynstr.2025.100727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/04/2025] [Accepted: 04/19/2025] [Indexed: 06/19/2025] Open
Abstract
Post-Traumatic Stress Disorder (PTSD) is a debilitating condition in which a traumatic experience triggers symptoms related to re-experiencing, avoidance, arousal, and mood dysregulation. PTSD negatively impacts 6 % of people during their lifetime, with women being disproportionally affected and exhibiting different, more severe symptoms than men. Despite this widespread impact, the molecular mechanisms underlying PTSD and its sex differences remain poorly understood. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) is a neuropeptide which participates in fine-tuning circuitry throughout the brain and has been associated with PTSD in humans, especially in women. Here, we use Single Prolonged Stress (SPS), an animal model of PTSD, to explore the roles of PACAP and sex in PTSD-like behaviors. Specifically, a PACAP agonist or antagonist was infused into the infralimbic (IL) prefrontal cortex, a region key to regulating fear- and anxiety-related behaviors, prior to SPS in male and female rats. Rats were then tested in open field/novel object, elevated plus maze, and social interaction. Utilizing a behavioral indexing method, we were able to uncover SPS effects in PTSD-related behavioral domains that were differentially impacted by PACAP manipulations in males and females. While both sexes exhibited increased threat avoidance and decreased threat assessment following SPS, females increased sociability while males decreased sociability. Males also appeared to be protected by IL PACAP antagonism while female SPS phenotypes were exacerbated by IL PACAP agonism. Furthermore, RNAscope revealed that PACAP in the prefrontal cortex responds differently to SPS in males and females. Together, these findings suggest complex relationships between SPS, sex, and IL PACAP which may have important implications for treating PTSD in men and women.
Collapse
Affiliation(s)
- Marissa A. Smail
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, United States
- Department of Psychology, Ohio State University, Columbus, OH, United States
| | - Evelin M. Cotella
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
- Department of Functional and Systems Neuroscience, Instituto Mercedes y Martin Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Susan E. Martelle
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
- RAI Services Company, Winston-Salem, NC, United States
| | - James B. Chambers
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Ria K. Parikh
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
- Program in Neuroscience, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Christine E. Moore
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Ben A. Packard
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Nawshaba Nawreen
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
- Eli Lilli and Company, Indianapolis, IN, United States
| | - Rachel D. Moloney
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - James P. Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
- Veterans Affairs Medical Center, Cincinnati, OH, United States
- Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
22
|
Zhang G, Lian Y, Li Q, Zhou S, Zhang L, Chen L, Tang J, Liu H, Li N, Pan Q, Gu Y, Lin N, Wang H, Wang X, Guo J, Zhang W, Jin Z, Xu B, Su X, Lin M, Han Q, Qin J. Vagal pathway activation links chronic stress to decline in intestinal stem cell function. Cell Stem Cell 2025; 32:778-794.e10. [PMID: 40120585 DOI: 10.1016/j.stem.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/11/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
Chronic stress adversely affects intestinal health, but the specific neural pathways linking the brain to intestinal tissue are not fully understood. Here, we show that chronic stress-induced activation of the central amygdala-dorsal motor nucleus of the vagus (CeA-DMV) pathway accelerates premature aging and impairs the stemness of intestinal stem cells (ISCs). This pathway influences ISC function independently of the microbiota, the hypothalamic-pituitary-adrenal (HPA) axis, the immune response, and the sympathetic nervous system (SNS). Under chronic stress, DMV-mediated vagal activation prompts cholinergic enteric neurons to release acetylcholine (ACh), which engages ISCs via the M3 muscarinic acetylcholine receptor (CHRM3). This interaction activates the p38 mitogen-activated protein kinase (MAPK) pathway, triggering growth arrest and mitochondrial fragmentation, thereby accelerating an aging-like decline in ISCs. Together, our findings provide insights into an alternative neural mechanism that links stress to intestinal dysfunction. Strategies targeting the DMV-associated vagal pathway represent potential therapeutic approaches for stress-induced intestinal diseases.
Collapse
Affiliation(s)
- Guoying Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yannan Lian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Shanghai 200032, China
| | - Shudi Zhou
- Department of Endocrinology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Lili Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Liting Chen
- Department of Emergency and Critical Disease, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junzhe Tang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Shanghai 200032, China
| | - Hailong Liu
- Department of General Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ni Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Qiang Pan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yongqiang Gu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Naiheng Lin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hanling Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xuege Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Jiacheng Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Wei Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zige Jin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Beitao Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xiao Su
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Moubin Lin
- Department of General Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Qi Han
- Department of Emergency and Critical Disease, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jun Qin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
23
|
Townsend LK, Wang D, Knuth CM, Fayyazi R, Mohammad A, Becker LJ, Tsakiridis EE, Desjardins EM, Patel Z, Valvano CM, Lu J, Payne AE, Itua O, Medak KD, Marko DM, Schertzer JD, Wright DC, Beaudette SM, Morrison KM, Carpentier AC, Blondin DP, MacPherson REK, McCall JG, Jeschke MG, Steinberg GR. GDF15 links adipose tissue lipolysis with anxiety. Nat Metab 2025; 7:1004-1017. [PMID: 40234625 PMCID: PMC12116386 DOI: 10.1038/s42255-025-01264-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/06/2025] [Indexed: 04/17/2025]
Abstract
Psychological stress changes both behaviour and metabolism to protect organisms. Adrenaline is an important driver of this response. Anxiety correlates with circulating free fatty acid levels and can be alleviated by a peripherally restricted β-blocker, suggesting a peripheral signal linking metabolism with behaviour. Here we show that adrenaline, the β3 agonist CL316,243 and acute restraint stress induce growth differentiation factor 15 (GDF15) secretion in white adipose tissue of mice. Genetic inhibition of adipose triglyceride lipase or genetic deletion of β-adrenergic receptors blocks β-adrenergic-induced increases in GDF15. Increases in circulating GDF15 require lipolysis-induced free fatty acid stimulation of M2-like macrophages within white adipose tissue. Anxiety-like behaviour elicited by adrenaline or restraint stress is eliminated in mice lacking the GDF15 receptor GFRAL. These data provide molecular insights into the mechanisms linking metabolism and behaviour and suggest that inhibition of GDF15-GFRAL signalling might reduce acute anxiety.
Collapse
Affiliation(s)
- Logan K Townsend
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Carly M Knuth
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Russta Fayyazi
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ahmad Mohammad
- Department of Health Science, Brock University, St. Catherines, Ontario, Canada
| | - Léa J Becker
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Evangelia E Tsakiridis
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Eric M Desjardins
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Zeel Patel
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Celina M Valvano
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Junfeng Lu
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alice E Payne
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ofure Itua
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Kyle D Medak
- Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | - Daniel M Marko
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - David C Wright
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shawn M Beaudette
- Department of Kinesiology, Brock University, St. Catherines, Ontario, Canada
| | - Katherine M Morrison
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - André C Carpentier
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Denis P Blondin
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Jordan G McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Marc G Jeschke
- David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
- Hamilton General Hospital, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
24
|
Arnold AR, Chassaing B, Lakhani K, Bergeron C, Shaughnessy EK, Rosenhauer AM, Stoehr MC, Horne B, Wilkinson T, Huhman KL. Consumption of dietary emulsifiers increases sensitivity to social stress in mice: A potential role for the COX molecular pathway. Horm Behav 2025; 172:105750. [PMID: 40311305 DOI: 10.1016/j.yhbeh.2025.105750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/19/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Chronic low-grade inflammation and exposure to stress are key contributing factors in the etiology and progression of many neuropsychiatric disorders. Dietary emulsifiers, such as carboxymethylcellulose (CMC) and polysorbate-80 (P80), are commonly added to processed foods and drinks and are classified by the Food and Drug Administration (FDA) as generally recognized as safe (GRAS). Recently, however, we and others have reported that these additives at translationally relevant doses cause low-grade intestinal inflammation, microbiota dysbiosis, and alterations in gene expression in brain areas that mediate behavioral and neuroendocrine responses to stress-provoking stimuli. METHODS To test whether emulsifier exposure sensitizes behavioral, hormonal, and neuronal responses to stress, C57BL/6 J male mice were given water +1 % emulsifier (CMC or P80) or water alone for 12 weeks after which they were exposed to social defeat stress. We previously found increased PTGS2 (COX-2) gene expression in the amygdala following emulsifier consumption. To determine whether inflammation, potentially through the COX pathway, is a potential mechanism driving emulsifier-induced increases in stress sensitivity, we administered the COX inhibitor aspirin (25 mg/kg/day) in conjunction with emulsifiers for the last six weeks of treatment. RESULTS In defeated mice, CMC increased circulating corticosterone, while both emulsifiers increased social avoidance behavior and altered defeat-induced c-Fos immunofluorescence in various brain regions. Moreover, behavioral and hormonal alterations were attenuated by aspirin. CONCLUSIONS These data demonstrate that ingestion of at least some dietary emulsifiers at concentrations analogous to those ingested by humans increases sensitivity to social stress in mice and that the COX pathway may be a mechanistic candidate by which emulsifier-induced increases in sensitivity to social stress occur.
Collapse
Affiliation(s)
- Amanda R Arnold
- Neuroscience Institute, Georgia State University, United States of America; Department of Psychiatry and Behavioral Sciences, Emory University, United States of America.
| | - Benoit Chassaing
- Neuroscience Institute, Georgia State University, United States of America; Microbiome-Host Interactions, INSERM U1306, CNRS UMR6047, Institut Pasteur, Université Paris Cité, Paris, France; Mucosal microbiota in chronic inflammatory diseases, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
| | - Kiran Lakhani
- Neuroscience Institute, Georgia State University, United States of America
| | - Coralie Bergeron
- Neuroscience Institute, Georgia State University, United States of America
| | - Emma K Shaughnessy
- Neuroscience Institute, Georgia State University, United States of America
| | - Anna M Rosenhauer
- Neuroscience Institute, Georgia State University, United States of America
| | - Maura C Stoehr
- Neuroscience Institute, Georgia State University, United States of America
| | - Benjamin Horne
- Neuroscience Institute, Georgia State University, United States of America
| | - Tyler Wilkinson
- Department of Counseling, Mercer University, United States of America
| | - Kim L Huhman
- Neuroscience Institute, Georgia State University, United States of America
| |
Collapse
|
25
|
Ohira M, Kawagoe N, Kameyama C, Kondou Y, Igarashi M, Ueshiba H. Association of serum cortisol with insulin secretion and plasma aldosterone with insulin resistance in untreated type 2 diabetes: a cross-sectional study. Diabetol Metab Syndr 2025; 17:144. [PMID: 40296149 PMCID: PMC12036189 DOI: 10.1186/s13098-025-01706-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Insulin secretion and resistance are key pathophysiological factors in type 2 diabetes. However, only 55% of patients achieve long-term blood glucose treatment goals, highlighting the need to clarify the pathophysiology of type 2 diabetes. While cortisol and aldosterone levels have been linked to insulin secretion and resistance in participants without type 2 diabetes, their role in patients with type 2 diabetes remains unclear. In this study, we aimed to investigate the relationships among insulin secretion, insulin resistance, and cortisol or aldosterone levels in patients with untreated type 2 diabetes. METHODS We retrospectively reviewed 121 patients with untreated type 2 diabetes mellitus. We analyzed the relationships between various clinical parameters, including adrenal hormones, and insulin secretion (homeostatic model assessment [HOMA2-%B]) or insulin resistance (HOMA2-IR). Multiple regression analysis was performed to identify parameters associated with HOMA2-%B or HOMA2-IR. RESULTS Spearman's rank correlation coefficient revealed that body weight (BW); body mass index (BMI); estimated glomerular filtration rate; and serum creatinine, uric acid, total cholesterol, high-density lipoprotein cholesterol (HDL-C), sodium, potassium, chloride, fasting blood glucose (FBG), glycated hemoglobin (HbA1c), serum C-peptide, and cortisol levels were significantly correlated with HOMA2-%B. Similarly, BW, BMI, aspartate transaminase levels, alanine transaminase (ALT) levels, triglyceride levels, HDL-C levels, FBG levels, serum C-peptide levels, renin activity, and plasma aldosterone concentration (PAC) were significantly correlated with HOMA2-IR. Multiple regression analysis revealed BMI, HbA1c levels, and cortisol levels as predictors of HOMA2-%B, whereas ALT levels and the PAC were predictors of HOMA2-IR. CONCLUSION Serum cortisol levels are associated with insulin secretion, and the PAC is associated with insulin resistance in patients with untreated type 2 diabetes. These findings suggest that aldosterone blockade may represent a potential therapeutic approach for reducing insulin resistance in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Masahiro Ohira
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Toho University Ohashi Medical Center, 2-22-36 Ohashi, Meguro-ku, Tokyo, 153-8515, Japan.
| | - Naoyuki Kawagoe
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Toho University Ohashi Medical Center, 2-22-36 Ohashi, Meguro-ku, Tokyo, 153-8515, Japan
| | - Chisato Kameyama
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Toho University Ohashi Medical Center, 2-22-36 Ohashi, Meguro-ku, Tokyo, 153-8515, Japan
| | - Yuko Kondou
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Toho University Ohashi Medical Center, 2-22-36 Ohashi, Meguro-ku, Tokyo, 153-8515, Japan
| | - Madoka Igarashi
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Toho University Ohashi Medical Center, 2-22-36 Ohashi, Meguro-ku, Tokyo, 153-8515, Japan
| | - Hajime Ueshiba
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Toho University Ohashi Medical Center, 2-22-36 Ohashi, Meguro-ku, Tokyo, 153-8515, Japan
| |
Collapse
|
26
|
Ferreira DH, Ryan R, Smyth N, Clow A, Currow DC. The longitudinal impact of low-dose morphine on diurnal cortisol profiles in people with chronic breathlessness and chronic obstructive pulmonary disease (COPD): an exploratory study. Respir Res 2025; 26:156. [PMID: 40269943 PMCID: PMC12020152 DOI: 10.1186/s12931-025-03230-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
INTRODUCTION Stress activates the hypothalamic-pituitary-adrenal (HPA) axis of which cortisol is an end product. 'Allostatic load' is where systems including the HPA axis are exposed to high, cumulative, physiologic burdens (such as chronic breathlessness) leading to flatter diurnal cortisol slopes and poorer health outcomes. The aim of this hypothesis-generating study explored longitudinal changes in cortisol secretion and any associated changes in breathlessness after introducing regular, low dose morphine or placebo. METHODS This was an optional, hypothesis-generating sub-study embedded in a multi-site, randomised, double-blind, placebo-controlled trial (RCT) of regular, low-dose morphine for chronic breathlessness and chronic obstructive pulmonary disease. In a blinded dose-increment algorithm by week three, doses were 0 mg-32 mg. Participants in the RCT could elect to continue in a six-month blinded extension. This sub-study excluded people who used non-inhaled corticosteroids in the previous month or were on subcutaneous insulin. Participants collected saliva for cortisol assays for two days at baseline, and ends of weeks 1, 3 and 12 at 3,6 and 12 h after waking, generating sufficient data to calculate diurnal cortisol slopes and areas under the curve (AUC). Samples were analysed using ELISA. Correlations between diurnal cortisol profiles (slope and AUC) and a range of measures were explored. RESULTS Twenty mostly female former smokers were in this sub-study. At baseline and the end of week 1, one-way ANOVA between-group analyses showed no significant differences in the log-transformed cortisol slope or ln-AUC. There was a strong correlation between the age-adjusted Charlson Comorbidity Index (CCI) and ln-AUC (r=-0.70, p < 0.001) and moderate correlation with age (r=-0.43, p = 0.06). In the blinded extension study, there was a self-selecting blinded group (n = 7) all on active medication. Global impression of change (GIC) was highly correlated with the diurnal cortisol slope (rs = 0.98, p = 0.01), and with decrease in average breathlessness (r = 0.89, p = 0.04). DISCUSSION This hypothesis-generating study did not show a relationship between the diurnal cortisol profile and morphine in people with chronic breathlessness and COPD. For the sub-group still on study at 12weeks, the cortisol curves became steeper as average breathlessness decreased and as global impression of change (GIC) improved, suggesting that reducing breathlessness may potentially positively impact the HPA axis in a sub-group of people. TRIAL REGISTRATION Registration Number NCT02720822 date registered 28/03/2016.
Collapse
Affiliation(s)
- Diana H Ferreira
- Graduate School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Richella Ryan
- Arthur Rank Hospice Charity, Cherry Hinton Road, Shelford Bottom, CB22 3FB, Cambridge, UK
| | - Nina Smyth
- University of Westminster, Regent St, W1B 2HW, London, UK
| | - Angela Clow
- University of Westminster, Regent St, W1B 2HW, London, UK
| | - David C Currow
- Faculty of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| |
Collapse
|
27
|
Fouhy LE, Mangano KM, Zhang X, Dawson-Hughes B, Cornell DJ, Tucker KL, Noel SE. Association between urinary catecholamines and glucocorticoids and bone mineral density and osteoporosis in Puerto Rican adults. J Bone Miner Res 2025; 40:500-510. [PMID: 39893568 DOI: 10.1093/jbmr/zjaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/14/2024] [Accepted: 11/23/2024] [Indexed: 02/04/2025]
Abstract
Chronic stress leads to elevated stress hormones, which may be linked to bone breakdown. Puerto Rican adults living on the US mainland have higher prevalence of stress than the general population, and higher and/or similar prevalence of osteoporosis compared with non-Hispanic Whites. The role of stress on bone remains unclear and may be modified by diet. A Dietary Approaches to Stop Hypertension (DASH) pattern, as a measure of dietary quality, was most protective for bone outcomes among Puerto Ricans. In this cross-sectional study, 958 Boston Puerto Rican Health Study participants were included (aged: 59.9 ± 7.6 yr). Stress markers (epinephrine, norepinephrine, cortisol) were collected via 12-h urine samples and elevated concentrations were categorized using sex-specific cutoffs. BMD was assessed via DXA. Analysis of covariance models with least squares means were used to test differences in mean BMD between participants with elevated and non-elevated stress markers. Multivariable logistic regression examined associations between stress markers and osteoporosis in postmenopausal females and males. Models were adjusted for age, height, smoking, alcohol use, education, glucocorticoid use, and diabetes. Higher urinary epinephrine was associated with lower BMD at the LS (p = .012), FN (p = .005), trochanter (p < .001), and TH (p < .001) in Puerto Rican adults, and with higher odds of osteoporosis among males (odds ratio = 4.01 [95%CI: 1.11, 14.5], p = .03). An interaction between DASH and norepinephrine was noted for postmenopausal females at the LS. No associations were noted for norepinephrine or cortisol (p > .11), although higher urinary norepinephrine was associated with lower LS BMD in postmenopausal females not taking estrogen, with lower adherence to DASH (p = .03). Higher urinary epinephrine and norepinephrine were associated with poorer bone outcomes in Puerto Rican adults, in a sex-specific manner, warranting future longitudinal studies to clarify associations. Dietary quality may moderate these associations.
Collapse
Affiliation(s)
- Liam E Fouhy
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
- Center for Population Health, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | - Kelsey M Mangano
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
- Center for Population Health, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | - Xiyuan Zhang
- Center for Population Health, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | - Bess Dawson-Hughes
- Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, United States
| | - David J Cornell
- Department of Physical Therapy and Kinesiology, University of Massachusetts Lowell, Lowell, MA 01851, United States
- Center for Population Health, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | - Katherine L Tucker
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
- Center for Population Health, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | - Sabrina E Noel
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
- Center for Population Health, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States
| |
Collapse
|
28
|
Adam O, Perret M, Simon L, Dondé C, Raverot V, Vallet W, Mondino M, Brunelin J. Prefrontal cortex stimulation prevents stress-induced HPA axis reactivity in people at familial risk of schizophrenia. Eur Psychiatry 2025; 68:e55. [PMID: 40254409 PMCID: PMC12090024 DOI: 10.1192/j.eurpsy.2025.2455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/24/2025] [Accepted: 03/30/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Schizophrenia is a multifactorial disorder with a range of risk factors. Dysregulation in the systems involved in the stress response is a key component of its pathophysiology. Individuals at risk of developing schizophrenia exhibit hyperreactivity to stress and altered cognitive performance, both known as vulnerability markers. This study aims to determine whether stimulation of the prefrontal cortex can reduce reactivity to stress in unaffected siblings of patients with schizophrenia. METHODS In a randomized, sham-controlled trial, 27 participants were assigned to receive either active (n = 14) or sham (n = 13) transcranial direct current stimulation (tDCS) over the prefrontal cortex for 30 min during exposure to an acute stressor. The stress response was measured biologically, via salivary cortisol levels, and cognitively, through a reality monitoring task, which serves as an intermediate cognitive vulnerability marker. RESULTS In contrast to the sham condition, active stimulation significantly reduced cortisol release in response to stress (F(9,216) = 1.972; p = 0.04) and prevented stress-induced impairment in reality monitoring (F(1,23) = 9.954; p = 0.004). CONCLUSIONS These findings suggest that tDCS should be a promising tool for reducing stress-induced biological and cognitive reactivity in a population at risk of schizophrenia.
Collapse
Affiliation(s)
- Ondine Adam
- Le Vinatier Psychiatrie Universitaire Lyon Métropole, Bron, France
- CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, PsyR2 Team, Université Claude Bernard Lyon 1, Bron, France
| | - Mélanie Perret
- Le Vinatier Psychiatrie Universitaire Lyon Métropole, Bron, France
- CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, PsyR2 Team, Université Claude Bernard Lyon 1, Bron, France
| | - Louis Simon
- Le Vinatier Psychiatrie Universitaire Lyon Métropole, Bron, France
- CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, PsyR2 Team, Université Claude Bernard Lyon 1, Bron, France
- Psychiatric Emergency Service, Hospices Civils de Lyon, Lyon, France
| | - Clément Dondé
- Université Grenoble Alpes, Grenoble, France
- INSERM U1216, Grenoble, France
- Psychiatry Department, CHU Grenoble Alpes, Grenoble, France
- Psychiatry Department, Centre Hospitalier Alpes-Isère, Saint-Egrève, France
| | - Véronique Raverot
- CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028, WAKING Team, Université Claude Bernard Lyon 1, Bron, France
- Centre de biologie et de pathologie Est, Hospices Civils de Lyon, Groupement Hospitalier Est, LBMMS, Lyon, France
| | - William Vallet
- Le Vinatier Psychiatrie Universitaire Lyon Métropole, Bron, France
- CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, PsyR2 Team, Université Claude Bernard Lyon 1, Bron, France
| | - Marine Mondino
- Le Vinatier Psychiatrie Universitaire Lyon Métropole, Bron, France
- CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, PsyR2 Team, Université Claude Bernard Lyon 1, Bron, France
| | - Jérôme Brunelin
- Le Vinatier Psychiatrie Universitaire Lyon Métropole, Bron, France
- CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, PsyR2 Team, Université Claude Bernard Lyon 1, Bron, France
| |
Collapse
|
29
|
Zhang X, Wang H, Kilpatrick LA, Dong TS, Gee GC, Beltran-Sanchez H, Wang MC, Vaughan A, Church A. Connectome modeling of discrimination exposure: Impact on your social brain and psychological symptoms. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111366. [PMID: 40239889 DOI: 10.1016/j.pnpbp.2025.111366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/22/2025] [Accepted: 04/12/2025] [Indexed: 04/18/2025]
Abstract
Discrimination is a social stressor that is associated with adverse health outcomes, but the underlying neural mechanisms remain unclear. The fusiform, including the fusiform face area (FFA) plays a critical role in face perception especially regarding hostile faces during discrimination exposure; and are key regions involved in social cognition. We compared resting-state spontaneous activity and connectivity of the fusiform and FFA, between 153 individuals (110 women) with high (N = 73) and low (N = 80) levels of discrimination (measured by the Everyday Discrimination Scale) and evaluated the relationships of these brain signatures with psychological outcomes and stress-related neurotransmitters. Discrimination-related group differences showed altered fusiform signal fluctuation dynamics (Hurst exponent) and connectivity. These alterations predicted discrimination experiences and correlated with anxiety, depression, and cognitive difficulties. A molecular architecture analysis using cross-modal spatial correlation of brain signatures and nuclear imaging derived estimates of stress-related neurotransmitters demonstrated overlap between discrimination-related connectivity and dopamine, serotonin, gamma-aminobutyric acid (GABA), and acetylcholine. Discrimination exposure associated with alterations in the fusiform and face processing area may reflect enhanced baseline preparedness and vigilance towards facial stimuli and decreased top-down regulation of potential threats. These brain alterations may contribute to increased vulnerability for the development of mental health symptoms, demonstrating clinical relevance of social cognition in stressful interpersonal relationships.
Collapse
Affiliation(s)
- Xiaobei Zhang
- G. Oppenheimer Center for Neurobiology of Stress & Resilience, at UCLA, United States of America; Vatche and Tamar Manoukian Division of Digestive Diseases, at UCLA, United States of America; David Geffen School of Medicine at UCLA, United States of America; University of California, Los Angeles, United States of America; UCLA Goodman-Luskin Microbiome Center, United States of America
| | - Hao Wang
- G. Oppenheimer Center for Neurobiology of Stress & Resilience, at UCLA, United States of America; State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, China
| | - Lisa A Kilpatrick
- G. Oppenheimer Center for Neurobiology of Stress & Resilience, at UCLA, United States of America; Vatche and Tamar Manoukian Division of Digestive Diseases, at UCLA, United States of America; David Geffen School of Medicine at UCLA, United States of America; University of California, Los Angeles, United States of America; UCLA Goodman-Luskin Microbiome Center, United States of America
| | - Tien S Dong
- G. Oppenheimer Center for Neurobiology of Stress & Resilience, at UCLA, United States of America; Vatche and Tamar Manoukian Division of Digestive Diseases, at UCLA, United States of America; David Geffen School of Medicine at UCLA, United States of America; University of California, Los Angeles, United States of America; UCLA Goodman-Luskin Microbiome Center, United States of America
| | - Gilbert C Gee
- Department of Community Health Sciences Fielding School of Public Health, United States of America; California Center for Population Research, UCLA, United States of America
| | - Hiram Beltran-Sanchez
- Department of Community Health Sciences Fielding School of Public Health, United States of America; California Center for Population Research, UCLA, United States of America
| | - May C Wang
- Department of Community Health Sciences Fielding School of Public Health, United States of America
| | - Allison Vaughan
- G. Oppenheimer Center for Neurobiology of Stress & Resilience, at UCLA, United States of America; Vatche and Tamar Manoukian Division of Digestive Diseases, at UCLA, United States of America; David Geffen School of Medicine at UCLA, United States of America; University of California, Los Angeles, United States of America; UCLA Goodman-Luskin Microbiome Center, United States of America
| | - Arpana Church
- G. Oppenheimer Center for Neurobiology of Stress & Resilience, at UCLA, United States of America; Vatche and Tamar Manoukian Division of Digestive Diseases, at UCLA, United States of America; David Geffen School of Medicine at UCLA, United States of America; University of California, Los Angeles, United States of America; UCLA Goodman-Luskin Microbiome Center, United States of America.
| |
Collapse
|
30
|
Richardson RS, Kryszak LA, Vendruscolo JCM, Koob GF, Leggio L, Vendruscolo LF. Evidence for independent actions of the CRF and ghrelin systems in binge-like alcohol drinking in mice. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111341. [PMID: 40139339 PMCID: PMC12043401 DOI: 10.1016/j.pnpbp.2025.111341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/16/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Alcohol use disorder (AUD) and binge drinking are highly prevalent public health issues. Both ghrelin and corticotrophin-releasing factor (CRF) drive stress responses and alcohol drinking. Despite evidence of a relationship between the ghrelin and CRF systems, their potential interaction in modulating alcohol drinking is unclear. We tested the effect of a brain-penetrant CRF1 receptor antagonist (R121919) and a peripherally restricted nonselective CRF receptor antagonist (astressin) on plasma ghrelin levels. We also tested effects of R121919 and astressin alone and combined with the growth hormone secretagogue receptor (GHSR; the ghrelin receptor) antagonist JMV2959 and GHSR antagonist/inverse agonist PF-5190457 in a model of binge-like alcohol drinking in male and female C57BL/6 J mice. The intraperitoneal administration of R121919 but not astressin increased plasma ghrelin levels. R121919 but not astressin reduced binge-like alcohol drinking. CRF receptor antagonism had no effect on the ability of GHSR blockers to reduce alcohol drinking. No sex × drug treatment interactions were observed. These findings suggest that while both CRF receptor antagonism and GHSR antagonism reduce alcohol drinking, these two pharmacological approaches may not interact to mediate binge-like alcohol drinking in mice. Additionally, these results provide evidence that GHSR but not peripheral endogenous ghrelin may be key in driving binge-like alcohol drinking.
Collapse
Affiliation(s)
- Rani S Richardson
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA; Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA; Stress and Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA; University of North Carolina School of Medicine MD/PhD Program, University of North Carolina, Chapel Hill, NC, USA; Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Lindsay A Kryszak
- Translational Analytical Core, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA; Translational Analytical Core, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA; Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA; Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA..
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
31
|
Abd-Elmonsif NM, Gamal S, Barsoom SA. Chronic stress and depression impact on tongue and major sublingual gland histology and the potential protective role of Thymus vulgaris: An animal study. Arch Oral Biol 2025; 172:106182. [PMID: 39864188 DOI: 10.1016/j.archoralbio.2025.106182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/12/2024] [Accepted: 01/18/2025] [Indexed: 01/28/2025]
Abstract
OBJECTIVES Reporting the histological effects of chronic stress on certain oral tissues, as well as the capacity of Thymus vulgaris (thyme) to protect tissues from stress and link both serum cortisol and serotonin levels. METHODS 30 rats were randomly divided into a trio of groups: normal control (no treatment), stress group (chronic stress without treatment), and treatment group (chronic stress treated with thyme at a dose of 200 mg/kg BW orally via needle gavage daily for 21 days). At the end of the experiment, tongues and major sublingual glands (SLGs) were surgically removed and processed for histological and histochemical studies. Blood samples were taken shortly before scarification for the biochemical study of cortisol and serotonin serum levels. RESULTS Examination of tongue and SLG sections of the stress group indicated significant alterations in histology and changes in SLG secretion. An examination of tongue and SLG histological sections of the thyme-treated group are showed an improvement. Chronic stress raises cortisol serum levels and lowers serotonin serum levels. CONCLUSIONS Chronic stress causes alteration of the tongue and major SLG histology, as well as changes in SLG secretion. Thyme may protect tissues from stress, and there is a relation between cortisol and serotonin levels.
Collapse
Affiliation(s)
- Nehad M Abd-Elmonsif
- Department of Oral Biology, Faculty of Oral and Dental Medicine, Future University in Egypt, Cairo, Egypt.
| | - Sherif Gamal
- Research Labs Supervisor, Faculty of pharmacy, Future University in Egypt, Cairo, Egypt
| | | |
Collapse
|
32
|
Becker L, Martin T, Rohleder N, Nieding G, Wannagat W. Physiological stress responses to digital single- and multitasking demands in younger and older adults. Psychoneuroendocrinology 2025; 174:107376. [PMID: 39893951 DOI: 10.1016/j.psyneuen.2025.107376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Differences in multitasking (MT) performance have been found across the lifespan. Little is known about physiological stress responses to MT demands in older people. In our study, 106 younger (22.8 ± 3.9 years, 75.5 % female) and 113 older adults (70.9 ± 5.1 years, 70.8 % female) participated and were assigned to either a single-tasking (ST) or an MT condition. Physiological stress responses of the Autonomic Nervous System (ANS), and the hypothalamic-pituitary adrenal (HPA) axis were assessed. Task 1 was a sentence verification task, which was interrupted by push notifications (task 2) in the MT condition. Participants answered questions that assessed their comprehension of the notification content afterwards. Performance was lower in the MT condition in the older participants in task 1. All participants perceived stress during both tasks. Autonomic responses (i.e., for heart rate) were found for the MT condition in the older participants. No differences in physiological stress responses were found for the further autonomic measures. Cortisol levels decreased throughout the session for all participants. Our results confirm that MT performance decreases in older age. This may be associated with stronger ANS responses during MT in older than in younger people. Our findings partially support the specificity hypothesis, i.e., that cognitive stressors specifically initiate responses of the ANS, but no responses of the HPA axis. More research with more demanding tasks and the inclusion of further factors such as experience with MT and educational level is needed.
Collapse
Affiliation(s)
- Linda Becker
- Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Department of Psychology, Health Psychology, Nägelsbachstr. 49a, Erlangen 91052, Germany.
| | - Tamara Martin
- Julius-Maximilians-Universität Würzburg, Department of Psychology, Developmental Psychology, Röntgenring 10, Würzburg 97070, Germany
| | - Nicolas Rohleder
- Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Department of Psychology, Health Psychology, Nägelsbachstr. 49a, Erlangen 91052, Germany
| | - Gerhild Nieding
- Julius-Maximilians-Universität Würzburg, Department of Psychology, Developmental Psychology, Röntgenring 10, Würzburg 97070, Germany
| | - Wienke Wannagat
- Julius-Maximilians-Universität Würzburg, Department of Psychology, Developmental Psychology, Röntgenring 10, Würzburg 97070, Germany; Dr. Herbert Brause Center for Media Literacy, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg 97074, Germany
| |
Collapse
|
33
|
Kogler L, Wang R, Luther T, Hofer A, Frajo-Apor B, Derntl B. Cortisol in schizophrenia spectrum disorders: A comprehensive meta-analysis. Front Neuroendocrinol 2025; 77:101186. [PMID: 39986355 DOI: 10.1016/j.yfrne.2025.101186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/10/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Schizophrenia spectrum disorders (SSD) are characterized by alterations in cortisol levels across various parameters, including stress reactivity, hair cortisol, and baseline levels, which may be influenced by antipsychotic treatment. To provide a comprehensive overview of cortisol dysregulation in SSD, we conducted meta-analyses assessing (1) the effects of antipsychotic treatment in SSD patients, and additionally comparing cortisol in SSD patients versus healthy controls (HC) (2) following stress induction (metabolic, physiological, psychological stressors), (3) in hair and (4) baseline levels. Systematic literature searches in PubMed, Web of Science, and PsycINFO (November 2024) identified 121 studies (9049 SSD patients) for inclusion. Meta-analytic results revealed that antipsychotic treatment significantly reduced cortisol levels in SSD (k = 16, g = -0.480, 95 % CI [-0.818, -0.142], p = 0.005). Additionally, compared to HC, SSD was associated with reduced cortisol suppression following dexamethasone exposure (k = 9, g = 0.299, 95 % CI [0.091, 0.507], p = 0.005) and with elevated baseline cortisol levels in the morning (k = 71, g = 0.38, 95 % CI [0.210, 0.546], p < 0.001) and evening (k = 11, g = 0.368, 95 % CI [0.076, 0.661], p = 0.014). However, there were no significant group differences in afternoon baseline cortisol, hair cortisol or cortisol reactivity to stress (p > 0.05). These findings offer a detailed understanding of cortisol alterations in SSD and improve our understanding of HPA axis dysregulation in SSD.
Collapse
Affiliation(s)
- Lydia Kogler
- Department of Psychiatry and Psychotherapy, Tübingen Centre for Mental Health (TüCMH), Medical Faculty, University of Tübingen, Calwerstrasse 14, 72076 Tübingen, Germany; German Center for Mental Health (DZPG) Partner Site Tübingen 72076 Tübingen, Germany.
| | - Rui Wang
- Department of Psychiatry and Psychotherapy, Tübingen Centre for Mental Health (TüCMH), Medical Faculty, University of Tübingen, Calwerstrasse 14, 72076 Tübingen, Germany
| | - Teresa Luther
- Leibniz-Institut für Wissensmedien, Knowledge Construction Lab, Schleichstraße 6, 72076 Tübingen, Germany
| | - Alex Hofer
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Division of Psychiatry I, Medical University Innsbruck, Innsbruck, Austria
| | - Beatrice Frajo-Apor
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Division of Psychiatry I, Medical University Innsbruck, Innsbruck, Austria
| | - Birgit Derntl
- Department of Psychiatry and Psychotherapy, Tübingen Centre for Mental Health (TüCMH), Medical Faculty, University of Tübingen, Calwerstrasse 14, 72076 Tübingen, Germany; German Center for Mental Health (DZPG) Partner Site Tübingen 72076 Tübingen, Germany
| |
Collapse
|
34
|
Hasegawa M, Kunisawa K, Wulaer B, Kubota H, Kurahashi H, Sakata T, Ando H, Fujigaki S, Fujigaki H, Yamamoto Y, Nagai T, Saito K, Nabeshima T, Mouri A. Chronic stress induces behavioural changes through increased kynurenic acid by downregulation of kynurenine-3-monooxygenase with microglial decline. Br J Pharmacol 2025; 182:1466-1486. [PMID: 39658392 DOI: 10.1111/bph.17407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/06/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Alterations in tryptophan-kynurenine (TRP-KYN) pathway are implicated in major depressive disorder (MDD). α7 nicotinic acetylcholine (α7nACh) receptor regulates the hypothalamic-pituitary-adrenal (HPA) axis. We have shown that deficiency of kynurenine 3-monooxygenase (KMO) induces depression-like behaviour via kynurenic acid (KYNA; α7nACh antagonist). In this study, we investigated the involvement of the TRP-KYN pathway in stress-induced behavioural changes and the regulation of the HPA axis. EXPERIMENTAL APPROACH Mice were exposed to chronic unpredictable mild stress (CUMS) and subjected to behavioural tests. We measured TRP-KYN metabolites and the expression of their enzymes in the hippocampus. KMO heterozygous mice were used to investigate stress vulnerability. We also evaluated the effect of nicotine (s.c.) on CUMS-induced behavioural changes and an increase in serum corticosterone (CORT) concentration. KEY RESULTS CUMS decreased social interaction time but increased immobility time under tail suspension associated with increased serum corticosterone concentration. CUMS increased KYNA levels via KMO suppression with microglial decline in the hippocampus. Kmo+/- mice were vulnerable to stress: they exhibited social impairment and increased serum corticosterone concentration even after short-term CUMS. Nicotine attenuated CUMS-induced behavioural changes and increased serum corticosterone concentration by inhibiting the increase in corticotropin-releasing hormone. Methyllycaconitine (α7nACh antagonist) inhibited the attenuating effect of nicotine. CONCLUSIONS AND IMPLICATIONS CUMS-induced behavioural changes and the HPA axis dysregulation could be induced by the increased levels of KYNA via KMO suppression. KYNA plays an important role in the pathophysiology of MDD as an α7nACh antagonist. Therefore, α7nACh receptor is an attractive therapeutic target for MDD.
Collapse
Affiliation(s)
- Masaya Hasegawa
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Bolati Wulaer
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Medical Science, Aichi, Japan
| | - Hisayoshi Kubota
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Hitomi Kurahashi
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Takatoshi Sakata
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Honomi Ando
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Suwako Fujigaki
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Hidetsugu Fujigaki
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Yasuko Yamamoto
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
| | - Taku Nagai
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
| | - Kuniaki Saito
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Medical Science, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Toshitaka Nabeshima
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
- Laboratory of Health and Medical Science Innovation (HMSI), Fujita Health University Graduate School of Medical Science, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Medical Sciences, Aichi, Japan
- International Center for Brain Science (ICBS), Fujita Health University, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| |
Collapse
|
35
|
Wang Z, Chang X, Zhang C, Lan H, Huang M, Zhou B, Sun B. Beyond Aromas: Exploring the Development and Potential Applications of Electroencephalography in Olfactory Research-From General Scents to Food Flavor Science Frontiers. Annu Rev Food Sci Technol 2025; 16:147-170. [PMID: 39745932 DOI: 10.1146/annurev-food-110124-035308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Olfaction is crucial to our dietary choices and significantly influences our emotional and cognitive landscapes. Understanding the underlying neural mechanisms is pivotal, especially through the use of electroencephalography (EEG). This technology has strong temporal resolution, allowing it to capture the dynamics of neural responses to odors, bypassing the need for subjective interpretations. The application of EEG in food flavor research is still relatively new, but it has great potential. This review begins with an examination of general scent stimulation, charts the advances in using EEG to understand odor perception, and explores its future in food flavor science. By analyzing EEG's ability to detect distinct patterns and strengths in brain activity, we can elucidate the perceptual, affective, and cognitive frameworks associated with food odors. Event-related potentials and oscillatory activities, markers of central olfactory processing, provide insights into the neural architecture of olfaction. These markers are instrumental in assessing the influence of food odors on health, emotions, and decision-making processes. We argue that EEG's application in olfaction research holds considerable promise for the food industry to innovate products that are not only healthier but also more appealing, thereby promoting human well-being.
Collapse
Affiliation(s)
- Zhen Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, China; ,
| | - Xiaoyue Chang
- State Key Laboratory of Brain and Cognitive Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China;
| | - Chongyu Zhang
- TUM School of Engineering and Design, Technical University of Munich, Munich, Germany
| | - Haihui Lan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mingquan Huang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, China; ,
| | - Bin Zhou
- State Key Laboratory of Brain and Cognitive Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China;
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing, China; ,
| |
Collapse
|
36
|
Zheng L, Pang Q, Huang R, Xu H, Guo H, Gao C, Chen X, Wang Y, Cao Q, Gao Y, Gu Z, Wang Z, Luo C, Tao L, Wang T. Stress-mediated Activation of Ferroptosis, Pyroptosis, and Apoptosis Following Mild Traumatic Brain Injury Exacerbates Neurological Dysfunctions. Mol Neurobiol 2025; 62:4055-4075. [PMID: 39388040 DOI: 10.1007/s12035-024-04516-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
Nearly half of mild traumatic brain injury (mTBI) patients continue to experience residual neurological dysfunction, which may be attributed to exposure to stress. Ferroptosis, a newly discovered form of cell death, is increasingly recognized for its involvement in the pathophysiology of TBI. Understanding the mechanisms by which stress influences mTBI, particularly through ferroptosis, is crucial for the effective treatment and prevention of mTBI patients who are sensitive to stressful events. In our study, a mouse mTBI model was established. An acute restraint stress (RS) and a chronic unpredictable mild stress (CUMS) model then were applied to make acute and chronic stress, respectively. We found acute RS significantly delayed the recovery of reduced body weight and short-term motor dysfunctions and exacerbated cell insults and blood-brain barrier leakage caused by mTBI. Further studies revealed that acute RS exacerbates neuronal ferroptosis, pyroptosis, and apoptosis by promoting iron overloading in the neocortex following mTBI. Interestingly, the inhibition of ferroptosis with iron chelators, including deferoxamine and ciclopirox, reversed pyroptosis and apoptosis. Moreover, CUMS aggravated neurological dysfunctions (motor function, cognitive function, and anxiety-like behavior) and exacerbated brain lesion volume. CUMS also exacerbates ferroptosis, pyroptosis, and apoptosis by intensifying iron deposition, along with decreasing the expression of neuronal brain-derived neurotrophic factor and glucocorticoid receptor in the neocortex post mTBI. These effects were also mitigated by iron chelators. Our findings suggest that alleviating ferroptosis induced by iron deposition may represent a promising therapeutic approach for mTBI patients who have experienced stressful events.
Collapse
Affiliation(s)
| | | | | | - Heng Xu
- Soochow University, Suzhou, China
| | | | | | | | | | - Qun Cao
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Yuan Gao
- Soochow University, Suzhou, China
| | - Zhiya Gu
- Soochow University, Suzhou, China
| | | | | | | | - Tao Wang
- Soochow University, Suzhou, China.
| |
Collapse
|
37
|
Cheng M, Jiang Z, Yang J, Sun X, Song N, Du C, Luo Z, Zhang Z. The role of the neuroinflammation and stressors in premenstrual syndrome/premenstrual dysphoric disorder: a review. Front Endocrinol (Lausanne) 2025; 16:1561848. [PMID: 40225329 PMCID: PMC11985436 DOI: 10.3389/fendo.2025.1561848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD) are prevalent emotional disorders in females, characterized by cyclic variations in physiological stress responses and emotional symptoms that correspond with the menstrual cycle. Despite extensive research, the underlying causes of these disorders remain elusive. This review delves into the neurobiological mechanisms connecting stress-induced neuroinflammation with PMS/PMDD. Additionally, it traces the conceptual development and historical context of PMS/PMDD. The review further evaluates clinical evidence on the association between PMS/PMDD and stress, along with findings from both clinical and animal studies that link these disorders to inflammatory processes. Additionally, the neurobiological pathways by which inflammatory responses may play a role in the pathogenesis of PMS/PMDD were elucidated, including their interactions with the hypothalamic-pituitary-ovary (HPO) axis, serotonin-kynurenine (5-HT-KYN) system, GABAergic system, brain-derived neurotrophic factor (BDNF), hypothalamic-pituitary-adrena(HPA)axis and. Future research is encouraged to further investigate the pathogenesis of PMS/PMDD through the perspective of neuroinflammatory responses.
Collapse
Affiliation(s)
- Ming Cheng
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Zhaoshu Jiang
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Jie Yang
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xu Sun
- Research and Development Department, Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei, China
| | - Nan Song
- Research and Development Department, Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei, China
| | - Chunyu Du
- Research and Development Department, Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei, China
| | - Zhenliang Luo
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Zhen Zhang
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Research and Development Department, Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei, China
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
38
|
Meng Y, Li Y, Gu H, Chen Z, Cui X, Wang X. Androgen receptors in corticotropin-releasing hormone neurons mediate the sexual dimorphism in restraint-induced thymic atrophy. Proc Natl Acad Sci U S A 2025; 122:e2426107122. [PMID: 40106355 PMCID: PMC11962470 DOI: 10.1073/pnas.2426107122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Sexual dimorphism in immune responses is well documented, but the underlying mechanisms remain incompletely understood. Here, we identified a subset of corticotropin-releasing hormone (CRH) neurons that express androgen receptors (ARs) as key mediators of sex differences in restraint-induced immunosuppression. Mechanistically, androgens directly activate AR-positive CRH neurons, enhancing the hypothalamic-pituitary-adrenal axis activation. This results in elevated corticosterone levels in response to restraint stress, leading to increased immune cell apoptosis and immune organ atrophy in male mice. Conditional knockout of ARs in CRH neurons eliminated this sexual dimorphism, highlighting ARs in CRH neurons as pivotal regulators of sex-specific immune responses to stress.
Collapse
Affiliation(s)
- Yutong Meng
- National Institute of Biological Sciences, Beijing and Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Yaning Li
- National Institute of Biological Sciences, Beijing and Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Huating Gu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
| | - Ziyao Chen
- National Institute of Biological Sciences, Beijing and Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Xiaoyang Cui
- Zhili College, Tsinghua University, Beijing100084, China
| | - Xiaodong Wang
- National Institute of Biological Sciences, Beijing and Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| |
Collapse
|
39
|
Franco-Villanueva A, Ford NC, Morano RL, Packard BA, Baccei ML, Herman JP. Time-dependent Actions of Corticosterone on Infralimbic Cortex Pyramidal Neurons of Adult Male Rats. J Neurosci 2025; 45:e0867242025. [PMID: 40101963 PMCID: PMC12060656 DOI: 10.1523/jneurosci.0867-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
Responses to acute stress function to restore homeostasis. Hence, the study of neurophysiological responses to acute stress helps to understand mechanisms underlying adaptive coping in the face of environmental demands. The infralimbic medial prefrontal cortex (IL-mPFC) modulates the switch between behavioral coping styles, and acute stress enhances glutamatergic neurotransmission on mPFC projection neurons. However, the role of acute stress responses and stress hormones on the physiology of IL-mPFC projection neurons during adulthood remains underexplored. Here, we studied rapid and slow effects of acute corticosterone exposure on synaptic transmission and intrinsic membrane excitability in layer 5 pyramidal neurons of the IL (L5-IL PNs) in adult male rats using ex vivo whole-cell patch-clamp of mPFC slices. We report that corticosterone dynamically modulates the physiology of L5-IL PNs in a time-dependent manner. Specifically, corticosterone elicits a strong rapid shift of the excitatory-inhibitory balance towards enhanced excitation with mineralocorticoid (MR) and glucocorticoid receptors (GR) playing complementarily roles. Also, corticosterone rapidly and transently decreases the firing rate of L5-IL PNs via GR. Moreover, acute stress or corticosterone slowly enhance glutamatergic neurotransmission via MR and GR without modulating inhibitory neurotransmission or intrinsic excitability of adult L5-IL PNs. Our findings highlight the potential relevance of corticosterone effects on L5-IL PNs to promote a homeostatic response in adult male rats. First, corticosterone rapidly attenuates IL intrinsic excitability during the rapid initial phase of the acute stress response. Later on, corticosterone slowly restores IL output function over time to promote adaptive executive responses when context changes.Significance statement Corticosterone modulates physiological processes during stress to support adaptation. However, acute effects of corticosterone on stress control networks remains underexplored. Here, we explored mechanisms underlying corticosterone regulation of the activity of stress regulatory neurons of the infralimbic cortex (IL). Stress levels of corticosterone rapidly shift the excitatory-inhibitory balance of synaptic transmission towards enhanced excitation while diminishing firing of IL excitatory long-range neurons (IL PNs). Slow, lasting effects of corticosterone primarily target excitatory synaptic activity. Synaptic actions of glucocorticoids are cooperatively mediated by the mineralocorticoid (MRs) and glucocorticoid receptors (GRs), whereas the transient reduction in firing relies on GR in IL PNs. Thus, corticosterone provides an adaptive signal that controls IL output over time, promoting adaptive responses to environmental context.
Collapse
Affiliation(s)
- Ana Franco-Villanueva
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, 45267, USA
| | - Neil C Ford
- Department of Anesthesiology, Pain Research Center, University of Cincinnati Medical Center, Cincinnati, Cincinnati, Ohio, 45267, USA
| | - Rachel L Morano
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, 45267, USA
| | - Benjamin A Packard
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, 45267, USA
| | - Mark L Baccei
- Department of Anesthesiology, Pain Research Center, University of Cincinnati Medical Center, Cincinnati, Cincinnati, Ohio, 45267, USA
| | - James P Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, 45267, USA
- Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, 45220, USA
| |
Collapse
|
40
|
Keter DL, Bialosky JE, Brochetti K, Courtney CA, Funabashi M, Karas S, Learman K, Cook CE. The mechanisms of manual therapy: A living review of systematic, narrative, and scoping reviews. PLoS One 2025; 20:e0319586. [PMID: 40100908 PMCID: PMC11918397 DOI: 10.1371/journal.pone.0319586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/09/2024] [Indexed: 03/20/2025] Open
Abstract
INTRODUCTION Treatment mechanisms are the underlying process or pathway through which a treatment influences the body. This includes molecular, cellular and physiological processes or pathways contributing to treatment effect. Manual therapy (MT) evokes complex mechanistic responses across body systems, interacting with the individual patient and context to promote a treatment response. Challenges arise as mechanistic studies are spread across multiple professions, settings and populations. The purpose of this review is to summarize treatment mechanisms that have been reported to occur with MT application. METHODS Four electronic databases were searched (Medline, CINAHL, Cochrane Library, and PEDro) for reviews investigating mechanistic responses which occur during/post application of MT. This review was registered a priori with PROSPERO (CRD42023444839). Methodological quality (AMSTAR-2) and risk of bias (ROBIS) were assessed for systematic and scoping reviews. Data were synthesized by mechanistic domain. RESULTS Sixty-two reviews were included. Systematic reviews (n = 35), narrative reviews (n = 24), and scoping reviews (n = 4) of asymptomatic (n = 37), symptomatic (n = 43), non-specified human subjects (n = 7) and animals (n = 7) were included. Reviews of moderate quality supported neurovascular, neurological, and neurotransmitter/neuropeptide changes. Reviews of low quality supported neuroimmunce, neuromuscular, and neuroendocrine changes. Reviews of critically low quality support biomechanical changes. CONCLUSIONS Findings support critically low to moderate quality evidence of complex multisystem mechanistic responses occurring with the application of MT. Results support peripheral, segmental spinal, and supraspinal mechanisms occurring with the application of MT, which can be measured directly or indirectly. The clinical value of these findings has not been well established. While MT has proven to be an effective intervention to treat conditions such as pain, the current body of literature leaves uncertainty as to 'why' MT interventions work, and future research should look to better define which mechanisms (or combinations of mechanisms) are mediators of clinical response.
Collapse
Affiliation(s)
- Damian L. Keter
- Physical Medicine and Rehabilitation Department, United States Department of Veterans Affairs, Cleveland, Ohio, United States of America
| | - Joel E. Bialosky
- Department of Physical Therapy, University of Florida, Gainesville, Florida, United States of America
- Brooks-PHHP Research Collaboration, Gainesville, Florida, United States of America
| | - Kevin Brochetti
- Physical Medicine and Rehabilitation Department, United States Department of Veterans Affairs, Cleveland, Ohio, United States of America
| | - Carol A. Courtney
- Department of Physical Therapy and Human Movement Sciences, Northwestern University, Chicago Illinois, United States of America
| | - Martha Funabashi
- Division of Research and Innovation, Canadian Memorial Chiropractic College, Toronto, Canada
- Department of Chiropractic, Université du Québec à Trois-Rivières, Trois-Rivières, Canada
- Research Center, Parker University, Dallas, Texas, United States of America
| | - Steve Karas
- Department of Physical Therapy, Chatham University, Pittsburgh, Pennsylvania, United States of America
| | - Kenneth Learman
- Department of Graduate Studies in Health and Rehabilitation Sciences, Youngstown State University, Youngstown, Ohio, United States of America
| | - Chad E. Cook
- Department of Orthopaedics, Duke University, Durham, North Carolina, United States of America
- Department of Population Health Sciences, Duke University, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
41
|
Raza ML. The stress-immune system axis: Exploring the interplay between stress and immunity. PROGRESS IN BRAIN RESEARCH 2025; 291:289-317. [PMID: 40222784 DOI: 10.1016/bs.pbr.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
The chapter talks about how our body and mind respond to stress and how it affects our immune system. Stress reactions, especially the fight-or-flight reaction, are helpful at first but can be harmful if they last too long. Long-term stress, caused by hormones like cortisol and adrenaline, weakens the immune system and makes people more likely to get sick. Important brain chemicals like serotonin and norepinephrine help control how our immune system works. Also, the connection between our gut and brain is an important way that mental health affects how our immune system functions. Getting older and experiencing stress early in life can affect how our immune system works. Inflammation caused by stress is connected to health issues like heart disease, depression, and autoimmune diseases. There are ways to manage stress, like being mindful and having support from friends, are important for keeping your immune system healthy and lessening harm caused by stress.
Collapse
Affiliation(s)
- Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| |
Collapse
|
42
|
Khundrakpam B, Segado M, Pazdera J, Gagnon Shaigetz V, Granek JA, Choudhury N. An Integrated Platform Combining Immersive Virtual Reality and Physiological Sensors for Systematic and Individualized Assessment of Stress Response (bWell): Design and Implementation Study. JMIR Form Res 2025; 9:e64492. [PMID: 40053709 PMCID: PMC11920663 DOI: 10.2196/64492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/06/2024] [Accepted: 11/27/2024] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Stress is a pervasive issue in modern society, manifesting in various forms such as emotional, physical, and work-related stress, each with distinct impacts on individuals and society. Traditional stress studies often rely on psychological, performance, or social tests; however, recently, immersive virtual reality (VR), which provides a sense of presence and natural interaction, offers the opportunity to simulate real-world tasks and stressors in controlled environments. Despite its potential, the use of VR to investigate the multifaceted manifestations of stress has not been thoroughly explored. OBJECTIVE This study aimed to explore the feasibility of using a VR-based platform, bWell, to elicit multifaceted stress responses and measure the resulting behavioral and physiological changes. Specifically, we aimed to design various VR stress exercises based on neurocardiac models to systematically test cardiac functioning within specific contexts of self-regulation (executive functioning, physical efforts, and emotional regulation). METHODS The development process adhered to guidelines for VR clinical trials and complex health interventions, encompassing 3 phases: preparation, development, and verification. The preparation phase involved a comprehensive literature review to establish links between stress, the heart, and the brain, leading to the formulation of a conceptual model based on the Neurovisceral Integration Model (NVIM) and Vagal Tank Theory (VTT). The development phase involved designing VR exercises targeting specific stressors and integrating physiological sensors such as photoplethysmography (PPG) and electromyography (EMG) to capture heart rate variability (HRV) and facial expressions. The verification phase, conducted with a small number of trials, aimed to design a study and implement a workflow for testing the feasibility, acceptability, and tolerability of the VR exercises. In addition, the potential for capturing physiological measures along with subjective ratings of stress for specific dimensions was assessed. RESULTS Verification trials demonstrated that the VR exercises were well tolerated, with negligible cybersickness and high user engagement. The different VR exercises successfully elicited the intended stress demands, along with the physiological responses. CONCLUSIONS The study presents a novel VR-based experimental setup that allows a systematic and individualized assessment of stress responses, paving the way for future research to identify features that confer stress resilience and help individuals manage stress effectively. While our conceptual model highlights the role of HRV in providing valuable insights into stress responses, future research will involve multivariate and machine learning analyses to predict individual stress responses based on comprehensive sensor data, including EMG and the VR-based behavioral data, ultimately guiding personalized stress management interventions.
Collapse
Affiliation(s)
| | - Melanie Segado
- National Research Council Canada, Boucherville, QC, Canada
| | - Jesse Pazdera
- National Research Council Canada, Boucherville, QC, Canada
| | | | - Joshua A Granek
- Defence Research and Development Canada, Toronto, ON, Canada
| | | |
Collapse
|
43
|
Valenza G, Matić Z, Catrambone V. The brain-heart axis: integrative cooperation of neural, mechanical and biochemical pathways. Nat Rev Cardiol 2025:10.1038/s41569-025-01140-3. [PMID: 40033035 DOI: 10.1038/s41569-025-01140-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/05/2025]
Abstract
The neural and cardiovascular systems are pivotal in regulating human physiological, cognitive and emotional states, constantly interacting through anatomical and functional connections referred to as the brain-heart axis. When this axis is dysfunctional, neurological conditions can lead to cardiovascular disorders and, conversely, cardiovascular dysfunction can substantially affect brain health. However, the mechanisms and fundamental physiological components of the brain-heart axis remain largely unknown. In this Review, we elucidate these components and identify three primary pathways: neural, mechanical and biochemical. The neural pathway involves the interaction between the autonomic nervous system and the central autonomic network in the brain. The mechanical pathway involves mechanoreceptors, particularly those expressing mechanosensitive Piezo protein channels, which relay crucial information about blood pressure through peripheral and cerebrovascular connections. The biochemical pathway comprises many endogenous compounds that are important mediators of neural and cardiovascular function. This multisystem perspective calls for the development of integrative approaches, leading to new clinical specialties in neurocardiology.
Collapse
Affiliation(s)
- Gaetano Valenza
- Neurocardiovascular Intelligence Lab, Department of Information Engineering & Research Center "E. Piaggio", University of Pisa, Pisa, Italy.
| | - Zoran Matić
- Neurocardiovascular Intelligence Lab, Department of Information Engineering & Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| | - Vincenzo Catrambone
- Neurocardiovascular Intelligence Lab, Department of Information Engineering & Research Center "E. Piaggio", University of Pisa, Pisa, Italy
| |
Collapse
|
44
|
Rodrigues B, Leitão RA, Santos M, Trofimov A, Silva M, Inácio ÂS, Abreu M, Nobre RJ, Costa J, Cardoso AL, Milosevic I, Peça J, Oliveiros B, Pereira de Almeida L, Pinheiro PS, Carvalho AL. MiR-186-5p inhibition restores synaptic transmission and neuronal network activity in a model of chronic stress. Mol Psychiatry 2025; 30:1034-1046. [PMID: 39237722 PMCID: PMC11835755 DOI: 10.1038/s41380-024-02715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
Chronic stress exerts profound negative effects on cognitive and emotional behaviours and is a major risk factor for the development of neuropsychiatric disorders. However, the molecular links between chronic stress and its deleterious effects on neuronal and synaptic function remain elusive. Here, using a combination of in vitro and in vivo approaches, we demonstrate that the upregulation of miR-186-5p triggered by chronic stress may be a key mediator of such changes, leading to synaptic dysfunction. Our results show that the expression levels of miR-186-5p are increased both in the prefrontal cortex (PFC) of mice exposed to chronic stress and in cortical neurons chronically exposed to dexamethasone. Additionally, viral overexpression of miR-186-5p in the PFC of naïve mice induces anxiety- and depressive-like behaviours. The upregulation of miR-186-5p through prolonged glucocorticoid receptor activation in vitro, or in a mouse model of chronic stress, differentially affects glutamatergic and GABAergic synaptic transmission, causing an imbalance in excitation/inhibition that leads to altered neuronal network activity. At glutamatergic synapses, we observed both a reduction in synaptic AMPARs and synaptic transmission, whereas GABAergic synaptic transmission was strengthened. These changes could be rescued in vitro by a miR-186-5p inhibitor. Overall, our results establish a novel molecular link between chronic glucocorticoid receptor activation, the upregulation of miR-186-5p and the synaptic changes induced by chronic stress, that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Beatriz Rodrigues
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Experimental Biology and Biomedicine Doctoral Programme, Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ricardo A Leitão
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Mónica Santos
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Alexander Trofimov
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Integrative Brain Function Neurobiology Lab, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, 197022, St. Petersburg, Russia
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 010000, Astana, Kazakhstan
| | - Mariline Silva
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Department of Applied Physics and Science for Life Laboratory (SciLifeLab), KTH Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Ângela S Inácio
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Mónica Abreu
- Multidisciplinary Institute of Aging, MIA Portugal, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Rui J Nobre
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- ViraVector, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Jéssica Costa
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Experimental Biology and Biomedicine Doctoral Programme, Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ana Luísa Cardoso
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ira Milosevic
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Multidisciplinary Institute of Aging, MIA Portugal, University of Coimbra, 3004-504, Coimbra, Portugal
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - João Peça
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Bárbara Oliveiros
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- iCRB-Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, 3000-548, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- ViraVector, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Paulo S Pinheiro
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal.
| | - Ana Luísa Carvalho
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal.
| |
Collapse
|
45
|
Khan Z, Messiri NE, Iqbal E, Hassan H, Tanweer MS, Sadia SR, Taj M, Zaidi U, Yusuf K, Syed NI, Zaidi M. On the role of epigenetic modifications of HPA axis in posttraumatic stress disorder and resilience. J Neurophysiol 2025; 133:742-759. [PMID: 39842807 DOI: 10.1152/jn.00345.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/09/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
Stress is a fundamental adaptive response that invokes amygdala and hypothalamus-pituitary-adrenal (HPA) axis along with other brain regions. Extreme or chronic stress, however, can result in a multitude of neuropsychiatric disorders, including anxiety, paranoia, bipolar disorder (BP), major depressive disorder (MDD), and posttraumatic stress disorder (PTSD). Despite widespread exposure to trauma (70.4%), the incidence of PTSD is relatively low (6.8%), suggesting that either individual susceptibility or adaptability driven by epigenetic and genetic mechanisms are likely at play. PTSD takes hold from exposure to traumatic events, such as death threats or severe abuse, with its severity being impacted by the magnitude of trauma, its frequency, and the nature. This comprehensive review examines how traumatic experiences and epigenetic modifications in hypothalamic-pituitary axis (HPA), such as DNA methylation, histone modifications, noncoding RNAs, and chromatin remodeling, are transmitted across generations, and impact genes such as FKBP prolyl isomerase 5 (FKBP5), nuclear receptor subfamily 3 group C member 1 (NR3C1), brain-derived neurotrophic factor (BDNF), and solute carrier family 6 member 4 (SLC6A4). It also provides a comprehensive overview on trauma reversal, resilience mechanisms, and pro-resilience factors such as histone acetyltransferases (HATs)/histone deacetylases (HDACs) ratio, dehydroepiandrosterone (DHEA)/cortisol ratio, testosterone levels, and neuropeptide Y, thus highlighting potential therapeutic approaches for trauma-related disorders. The studies highlighted here underscore the narrative, for the first time, that the examination and treatment of PTSD and other depressive disorders must invoke a multitude of approaches to seek out the most effective and personalized strategies. We also hope that the discussion emanating from this review will also inform government policies directed toward intergenerational trauma and PTSD.
Collapse
Affiliation(s)
- Zainab Khan
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nour El Messiri
- Department of Epidemiology, The University of Texas Health Science Center at Houston, Houston, Texas, United States
- Intergenerational Trauma Research Unit, Think for Actions, Calgary, Alberta, Canada
| | - Emann Iqbal
- Intergenerational Trauma Research Unit, Think for Actions, Calgary, Alberta, Canada
| | - Hadi Hassan
- Intergenerational Trauma Research Unit, Think for Actions, Calgary, Alberta, Canada
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Mohammad S Tanweer
- Intergenerational Trauma Research Unit, Think for Actions, Calgary, Alberta, Canada
| | - Syeda R Sadia
- Intergenerational Trauma Research Unit, Think for Actions, Calgary, Alberta, Canada
| | - Moizzuddin Taj
- Intergenerational Trauma Research Unit, Think for Actions, Calgary, Alberta, Canada
- Department of Mathematics and Statistics, University of Calgary, Calgary, Alberta, Canada
| | - Umar Zaidi
- Intergenerational Trauma Research Unit, Think for Actions, Calgary, Alberta, Canada
- Department of Natural Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Kamran Yusuf
- Section of Neonatology, Department of Pediatrics, School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Naweed I Syed
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Mukarram Zaidi
- Department of Family Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
46
|
Nashawi H, Foltz CT, Smail MA, Buesing DR, Herman JP, Ulrich-Lai YM. The impact of limited sucrose intake on perineuronal nets of parvalbumin interneurons in the basolateral amygdala: A potential role in stress resilience. Physiol Behav 2025; 290:114774. [PMID: 39631451 PMCID: PMC11789926 DOI: 10.1016/j.physbeh.2024.114774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Natural rewards like regular sucrose consumption can buffer physiological and behavioral stress responses, likely mediated, at least in part, by increased plasticity in parvalbumin-positive (PV+) interneurons in the basolateral amygdala (BLA). As PV+ interneuron plasticity is tightly regulated by specialized extracellular matrix structures called perineuronal nets (PNNs), this study investigated the impact of regular sucrose consumption vs. repetitive stress on the PNNs that surround PV+ interneurons in the BLA, as well as the number of glutamatergic (vGLUT1) and GABAergic (vGAT) appositions that PV+ cells receive. Male rats were given an established limited sucrose intake (LSI) feeding paradigm (vs. water-fed controls) and were co-exposed to a brief restraint stress (vs. no stress controls), daily for 14 days. Sucrose consumption increased the proportion of PV+ cells that were surrounded by PNNs, independent of stress exposure. PV+ cells with PNNs had more vGLUT1-positive and fewer vGAT-positive appositions compared to those lacking PNNs. Additionally, sucrose consumption increased the ratio of excitatory/inhibitory appositions onto PV+ cells, suggesting the possibility of elevated PV+ interneuron tone, leading to greater inhibition of the BLA's stress-excitatory output. These findings indicate that sucrose consumption influences PNN formation and structural plasticity on PV+ interneurons in the BLA, which has implications for understanding the neurological mechanisms underlying stress resilience by natural rewards.
Collapse
Affiliation(s)
- Houda Nashawi
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Corey T Foltz
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA
| | - Marissa A Smail
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Dana R Buesing
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA
| | - James P Herman
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA
| | - Yvonne M Ulrich-Lai
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
47
|
Golonka K, Gulla B, Kościelniak D, Darczuk D, Cienkosz-Stepanczak B, Bystrowska B, Fortuna D, Kozicz T, Krzyściak W. Sensory processing sensitivity in adult dental patients and its relation to perceived stress, cortisol, and serotonin secretion. Sci Rep 2025; 15:7328. [PMID: 40025052 PMCID: PMC11873274 DOI: 10.1038/s41598-025-90263-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/11/2025] [Indexed: 03/04/2025] Open
Abstract
Sensory processing sensitivity (SPS) is a biologically determined trait that influences how individuals respond to external and internal stimuli. A high level of SPS is characterized by three factors: increased emotional reactivity, heightened sensitivity to subtle stimuli, and greater susceptibility to overstimulation, all of which may impact well-being and health. This study examined the relationships between SPS, perceived stress, affect, and biochemical responses in adult dental patients (N = 157) on the day of a routine dental visit. Biochemical measures included morning cortisol and serotonin secretion (saliva samples), and cortisol concentration accumulated in recent months (hair sample). Perceived stress and negative and positive affect were assessed while patients waited for a dental procedure. The correlation analysis revealed that higher SPS level was associated with elevated hair cortisol and more negative affect. Cluster analyses tested SPS and its factors independently, revealing that individuals with higher SPS had higher cortisol levels in saliva and hair samples, as well as greater perceived stress and negative affect. Salivary serotonin levels showed varied relationships with different SPS factors, indicating the need to analyze SPS as a multidimensional construct. The results indicate that increased hair and salivary cortisol may be considered as biomarkers of SPS. In the context of patient-centered care, considering SPS levels may contribute to enhanced motivation for regular dental visits and improved treatment adherence.
Collapse
Affiliation(s)
- Krystyna Golonka
- Institute of Applied Psychology, Faculty of Management and Social Communication, Jagiellonian University, 30-348, Kraków, Poland.
| | - Bożena Gulla
- Institute of Applied Psychology, Faculty of Management and Social Communication, Jagiellonian University, 30-348, Kraków, Poland
| | - Dorota Kościelniak
- Department of Developmental Dentistry, Institute of Dentistry, Jagiellonian University Medical College, 31-155, Kraków, Poland
| | - Dagmara Darczuk
- Department of Periodontology, Prophylaxis and Oral Medicine, Institute of Dentistry, Jagiellonian University Medical College, 31-155, Kraków, Poland
| | - Beata Cienkosz-Stepanczak
- Laboratory of Anthropology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Kraków, 30-387, Poland
| | - Beata Bystrowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688, Kraków, Poland
| | - Dominika Fortuna
- Doctoral School in the Social Sciences, Jagiellonian University, 31-010, Kraków, Poland
| | - Tamas Kozicz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York, NY, 10029, USA
| | - Wirginia Krzyściak
- Department of Medical Diagnostics, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688, Kraków, Poland
| |
Collapse
|
48
|
Langer K, Wolf OT, Merz CJ, Jentsch VL. The effects of stress hormones on cognitive emotion regulation: A systematic review and integrative model. Neurosci Biobehav Rev 2025; 170:106040. [PMID: 39909150 DOI: 10.1016/j.neubiorev.2025.106040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
The experience of stress and the need to regulate emotions are pervasive in everyday life. Emotion regulation (ER) is particularly required under stress to facilitate successful adaptation and recovery. Importantly, a growing body of work has identified stress and ER deficits as transdiagnostic risk factors for psychopathology. This highlights the relevance of understanding how stress impacts ER to elucidate individual vulnerability to mental disorders. Stress alters cognitive and emotional functioning via stress hormones secreted by the two major stress systems: sympathetic nervous system and hypothalamus-pituitary adrenocortical axis. This review aims to compile and synthesize empirical studies in humans investigating the effects of acute stress and stress hormones on ER. A systematic literature search yielded 14 relevant studies, 11 investigating acute stress effects and 3 examining the influence of pharmacological cortisol elevations on ER. The results of the stress studies are mixed revealing either impairing, beneficial or no effects at all. Cortisol administration mostly facilitated ER attempts. Notably, we detected timing differences in measuring ER performance relative to stress exposure that potentially reconcile divergent findings. Here, we propose the PRESSURE model (Predominant Stress System Underpins Regulation of Emotions) postulating that the direction and magnitude of stress effects on ER depends on the relative predominance of one stress system over the other. Additionally, sex-stress hormone interactions, stimulus intensity and ER strategy are discussed as possible moderators. Finally, we highlight limitations in current research and provide recommendations for future studies that will further advance our understanding of the intricate relationship between stress and ER.
Collapse
Affiliation(s)
- Katja Langer
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Germany.
| | - Oliver T Wolf
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Germany
| | - Christian J Merz
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Germany
| | - Valerie L Jentsch
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Germany
| |
Collapse
|
49
|
Becker L, Sieber C, Rohleder N, Freiberger E, Kob R, Britting S. Physiological Stress in Safer Cycling in Older Age (SiFAr-Stress): A Randomized Controlled Trial. J Appl Gerontol 2025:7334648251316950. [PMID: 40008963 DOI: 10.1177/07334648251316950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
One possibility for maintaining mobility in older age is cycling. We investigated the impact of the multicomponent "Safer Cycling in Older Age" (SiFAr) intervention on psychological and physiological stress. Participants were 98 community-dwelling older adults (73.4 ± 5.4 years). Bedtime cortisol, hair cortisol concentrations, and C-reactive protein were measured before and after the 8-week SiFAr intervention and at follow-up. Additionally, acute stress responses were assessed during the second and seventh training sessions using salivary alpha-amylase and cortisol assessments. We found a decrease in acute perceived stress, anxiety, fear of falling, and uncertainty during the cycling trainings. Moreover, long-term perceived stress significantly decreased. No significant changes were found for any of the physiological stress measures. We conclude that cycling had a positive impact on perceived stress and wellbeing. Further research with more intense trainings is needed to fully understand the associations between cycling in older age and physiological stress.
Collapse
Affiliation(s)
- Linda Becker
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany
| | - Cornel Sieber
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany
- Kantonsspital Winterthur, Winterthur, Switzerland
| | - Nicolas Rohleder
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany
| | - Ellen Freiberger
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany
| | - Robert Kob
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany
| | - Sabine Britting
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany
| |
Collapse
|
50
|
Cheng Y, Yang R, Jia Y, Zhou Y, Yao Y, Shen C, Li D, Zeng R, Wan Z, Zhao Q, Jiang L, Liao X. The association of chronic pain, painkiller use, and potential mediators with liver fat content. Sci Rep 2025; 15:6688. [PMID: 39994347 PMCID: PMC11850618 DOI: 10.1038/s41598-025-89496-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Excessive accumulation of liver fat content (LFC) is a pathological manifestation of steatotic liver diseases. This study aims to investigate the relationship between chronic pain and LFC development. In the UK Biobank, chronic pain sites were collected via questionnaire, while LFC was measured by magnetic resonance imaging and quantified by Proton Density Fat Fraction (PDFF). During the median follow-up of 10.5 (4.0-17.8) years, in 39,437 individuals, neck/shoulder, back, stomach/abdominal, knee, and general pain achieved significant arithmetic means difference of 0.02, 0.02, 0.04, 0.02, and 0.15 in PDFF (P < 0.05) using multivariable linear regression models. There was a significant dose-effect for number of pain sites and PDFF (P < 0.001). Additionally, the link between pain sites and PDFF was much stronger in aspirin users than non-users, while steroids had the reverse effect (P for interaction < 0.05). C-reactive protein, sleep, diet, and depression were proved to mediated 8.41%, 13.3%, 6.6%, and 23.0% of the relationship, respectively. In conclusion, there were quantified differences in the relationship between chronic pain and LFC. For chronic pain patients with potential liver health issues, aspirin may be prioritized as an analgesic option due to its potential protective benefits, whereas steroid medications should be avoided.
Collapse
Affiliation(s)
- Yu Cheng
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Yang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Jia
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yiheng Zhou
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Yao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Can Shen
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dongze Li
- Department of Emergency Medicine, Disaster Medical Center, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zeng
- Department of Cardiology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhi Wan
- Department of Emergency Medicine, Disaster Medical Center, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Zhao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
- Teaching & Research Section of General Practice, The General Practice Medical Center, West China Hospital of Sichuan University, Chengdu, China
| | - Lihua Jiang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China.
- Teaching & Research Section of General Practice, The General Practice Medical Center, West China Hospital of Sichuan University, Chengdu, China.
- Department of health policy and management, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Xiaoyang Liao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China.
- Teaching & Research Section of General Practice, The General Practice Medical Center, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|