1
|
Shangase KB, Luvuno M, Mabandla M. Effects of combined postweaning social isolation and ketamine administration on schizophrenia-like behaviour in male Sprague Dawley rats. Behav Brain Res 2025; 476:115214. [PMID: 39182622 DOI: 10.1016/j.bbr.2024.115214] [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: 04/10/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
The pathophysiology behind negative and cognitive symptoms of schizophrenia is not well understood, thus limiting the effectiveness of treatment on these symptoms. Developing reliable animal model of schizophrenia is vital to advance our understanding on the neurobiological basis of the disorder. Double hit is used to refer to the use of two schizophrenia inducing interventions viz ketamine exposure and social isolation. In this study we aim to investigate the robustness of double hit model of schizophrenia in inducing negative and cognitive symptoms of schizophrenia. On postnatal day (PND) 23, thirty-two male Sprague Dawley rats were randomly grouped into four equal groups as follows: group housed + saline (GH), group housed + ketamine (GHK), isolated + saline (SI), and isolated + ketamine (SIK). A single ketamine dose (16 mg/kg) was administered 3 times a week for four weeks. Isolated animals were housed singly throughout the study. The following behavioural tests were carried out: elevated plus maze, three chamber social interaction, resident intruder tests, and novel object recognition (NOR). The SIK group exhibited high anxiety levels, with increased ACTH, corticosterone and norepinephrine concentration when compared to the other groups. The SIK animals also presented with reduced social interaction and decreased oxytocin concentration. SIK rats were more aggressive towards a juvenile intruder but had low testosterone concentration. The SIK group or double hit model showed impaired visual learning and memory and increased expression of proinflammatory cytokines. This suggest that the double hit model is more robust in inducing negative and cognitive symptoms of schizophrenia than each treatment alone.
Collapse
Affiliation(s)
- Khanyiso Bright Shangase
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa.
| | - Mluleki Luvuno
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Musa Mabandla
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
2
|
Perez-Caballero L, Guillot de Suduiraut I, Romero LR, Grosse J, Sandi C, Andero R. Corticosterone administration immediately after peripuberty stress exposure does not prevent protracted stress-induced behavioral alterations. Psychoneuroendocrinology 2024; 170:107164. [PMID: 39146600 DOI: 10.1016/j.psyneuen.2024.107164] [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: 02/27/2024] [Revised: 07/11/2024] [Accepted: 08/04/2024] [Indexed: 08/17/2024]
Abstract
Stress-related disorders are commonly associated with abnormalities in hypothalamic-pituitary-adrenal (HPA) axis activity. Preliminary studies with cortisol administration in the aftermath of trauma suggest that this HPA axis hormone can potentially prevent maladaptive behavioral and biological stress responses. However, the efficacy of glucocorticoid administration during the peripuberty period has not been tested yet, although this lifetime is a critical time window in brain development and is highly sensitive to the harmful effects of stress. To further examine the short and long-lasting impact of glucocorticoids treatment given during the post-peripubertal stress period, the present study utilized a rat model of peripubertal stress-induced psychopathology and animals were subjected to a battery of tests to assess anxiety-like behaviors, exploratory behavior and reactivity to novelty at late adolescence and sociability, anhedonia and stress coping behaviors at adulthood. All the experiments were performed in males and females to evaluate the potential behavioral sex differences. Overall, our results demonstrated that rats exposed to peripubertal stress show decreased sociability in adulthood without differences in anxiety and depression-like behaviors. Moreover, this study shows that the administration of corticosterone after stress exposure at peripuberty does not prevent stress-induced behavioral alterations. However, we observed that some stress-induced behavioural alterations and corticosterone responses are sex-specific. Thus, the data obtained highlight that delineating sex differences in stress-related studies may ultimately contribute to the development of effective therapeutic interventions for each sex.
Collapse
Affiliation(s)
- Laura Perez-Caballero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Isabelle Guillot de Suduiraut
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Synapsy Center for Neuroscience and Mental Health Research, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Leire R Romero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona 08193, Spain
| | - Jocelyn Grosse
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Synapsy Center for Neuroscience and Mental Health Research, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Synapsy Center for Neuroscience and Mental Health Research, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Raul Andero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; Centro de Investigación Biomédica En Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid 28029, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain; ICREA, Pg Lluís Companys 23, Barcelona 08010, Spain.
| |
Collapse
|
3
|
Amraoui J, Leclerc G, Jarlier M, Diaz J, Guler R, Demoly C, Verin C, Rey Dit Guzer S, Chalbos P, Moussion A, Taoum C, Neron M, Philibert L. Cardiac coherence and medical hypnosis: a feasibility study of a new combined approach for managing preoperative anxiety in patients with breast or gynaecological cancer. BJA OPEN 2024; 12:100309. [PMID: 39381542 PMCID: PMC11459624 DOI: 10.1016/j.bjao.2024.100309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/16/2024] [Indexed: 10/10/2024]
Abstract
Background Non-pharmaceutical approaches can help manage preoperative anxiety, but few studies have evaluated psychoeducational programmes, especially for cancer surgery. We assessed the feasibility of the COHErence Cardiaque (COHEC) programme where cardiac coherence and medical hypnosis are combined to manage preoperative anxiety in patients undergoing breast or gynaecological cancer surgical interventions (BGCSI). Methods Patients undergoing BGCSI were enrolled and followed a daily home programme with cardiac coherence and medical hypnosis sessions, starting 7 days before the procedure. The primary endpoint was optimal patient adherence (i.e. completion of ≥14 sessions). Secondary endpoints were anxiety levels, measured using the Visual Analogue Scale (VAS) and the Amsterdam Preoperative Anxiety and Information Scale (APAIS), satisfaction (EVAN-G), and quality of postoperative recovery (QoR-15). Results In total, 53 patients [mean age: 55 (34-82) yr] were included; 83.7% had breast cancer and 15.1% had gynaecological cancer. Optimal adherence was achieved by 64.2% (95% confidence interval: 49.8-76.9%) of the intention-to-treat population. Among the 43 patients who completed at least one session, exploratory analysis showed that anxiety on the day before (P=0.02) and the morning of the intervention (P=0.04) was decreased in patients with severe anxiety at baseline (VAS ≥70). The median VAS satisfaction score for the programme was 10 (4-10). Overall, 94% of patients were willing to include the COHEC programme in their daily routine. Conclusions The implementation of a psychoeducational programme combining cardiac coherence and medical hypnosis is feasible and might potentially help patients undergoing BGCSI to manage preoperative anxiety. A randomised trial is underway to assess the efficacy of the COHEC programme. Clinical trial registration NCT03981731.
Collapse
Affiliation(s)
- Jibba Amraoui
- Department of Anaesthesia, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Gilles Leclerc
- Department of Anaesthesia, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Marta Jarlier
- Biometrics Unit, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Jesus Diaz
- Department of Anaesthesia, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Ridvan Guler
- Department of Anaesthesia, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Clément Demoly
- Department of Anaesthesia, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Catherine Verin
- Department of Anaesthesia, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Sophie Rey Dit Guzer
- Department of Anaesthesia, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Patrick Chalbos
- Department of Clinical Research and Innovation, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Aurore Moussion
- Department of Clinical Research and Innovation, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Christophe Taoum
- Department of Surgical Oncology, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| | - Mathias Neron
- Department of Surgical Oncology, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
- INSERM U1194, Montpellier Cancer Research Institute (IRCM), University of Montpellier, Montpellier, France
| | - Laurent Philibert
- Department of Pharmacy, Montpellier Cancer Institute, University of Montpellier, Montpellier, France
| |
Collapse
|
4
|
Kremer TL, Chen J, Buhl A, Berhe O, Bilek E, Geiger LS, Ma R, Moessnang C, Reichert M, Reinhard I, Schwarz K, Schweiger JI, Streit F, Witt SH, Zang Z, Zhang X, Nöthen MM, Rietschel M, Ebner-Priemer UW, Schwarz E, Meyer-Lindenberg A, Braun U, Tost H. Multimodal Associations of FKBP5 Methylation With Emotion-Regulatory Brain Circuits. Biol Psychiatry 2024; 96:858-867. [PMID: 38460581 DOI: 10.1016/j.biopsych.2024.03.003] [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: 08/16/2023] [Revised: 02/02/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND Understanding the biological processes that underlie individual differences in emotion regulation and stress responsivity is a key challenge for translational neuroscience. The gene FKBP5 is a core regulator in molecular stress signaling that is implicated in the development of psychiatric disorders. However, it remains unclear how FKBP5 DNA methylation in peripheral blood is related to individual differences in measures of neural structure and function and their relevance to daily-life stress responsivity. METHODS Here, we characterized multimodal correlates of FKBP5 DNA methylation by combining epigenetic data with neuroimaging and ambulatory assessment in a sample of 395 healthy individuals. RESULTS First, we showed that FKBP5 demethylation as a psychiatric risk factor was related to an anxiety-associated reduction of gray matter volume in the ventromedial prefrontal cortex, a brain area that is involved in emotion regulation and mental health risk and resilience. This effect of epigenetic upregulation of FKBP5 on neuronal structure is more pronounced where FKBP5 is epigenetically downregulated at baseline. Leveraging 208 functional magnetic resonance imaging scans during a well-established emotion-processing task, we found that FKBP5 DNA methylation in peripheral blood was associated with functional differences in prefrontal-limbic circuits that modulate affective responsivity to daily stressors, which we measured using ecological momentary assessment in daily life. CONCLUSIONS Overall, we demonstrated how FKBP5 contributes to interindividual differences in neural and real-life affect regulation via structural and functional changes in prefrontal-limbic brain circuits.
Collapse
Affiliation(s)
- Thomas L Kremer
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Junfang Chen
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anais Buhl
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Oksana Berhe
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Edda Bilek
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Lena S Geiger
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ren Ma
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carolin Moessnang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Markus Reichert
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mental mHealth Lab, Chair of Applied Psychology, Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany; Department of eHealth and Sports Analytics, Ruhr University Bochum, Bochum, Germany
| | - Iris Reinhard
- Department of Biostatistics, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kristina Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Janina I Schweiger
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Fabian Streit
- DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H Witt
- DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Zhenxiang Zang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Xiaolong Zhang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany; Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Marcella Rietschel
- DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ulrich W Ebner-Priemer
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Mental mHealth Lab, Chair of Applied Psychology, Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Urs Braun
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Heike Tost
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany.
| |
Collapse
|
5
|
Alewel DI, Kodavanti UP. Neuroendocrine contribution to sex-related variations in adverse air pollution health effects. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2024; 27:287-314. [PMID: 39075643 DOI: 10.1080/10937404.2024.2383637] [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: 07/31/2024]
Abstract
Air pollution exposure is ranked as a leading environmental risk factor for not only cardiopulmonary diseases but also for systemic health ailments including diabetes, reproductive abnormalities, and neuropsychiatric disorders, likely mediated by central neural stress mechanisms. Current experimental evidence links many air pollution health outcomes with activation of neuroendocrine sympathetic-adrenal-medullary and hypothalamic-pituitary-adrenal (HPA) stress axes associated with resultant increases in adrenal-derived hormone levels acting as circulating mediators of multi-organ stress reactions. Epidemiological and experimental investigations also demonstrated sex-specific responses to air pollutant inhalation, which may be attributed to hormonal interactions within the stress and reproductive axes. Sex hormones (androgens and estrogens) interact with neuroendocrine functions to influence hypothalamic responses, subsequently augmenting stress-mediated metabolic and immune changes. These neurohormonal interactions may contribute to innate sex-specific responses to inhaled irritants, inducing differing individual susceptibility. The aim of this review was to: (1) examine neuroendocrine co-regulation of the HPA axis by gonadal hormones, (2) provide experimental evidence demonstrating sex-specific respiratory and systemic effects attributed to air pollutant inhalation exposure, and (3) postulate proposed mechanisms of stress and sex hormone interactions during air pollution-related stress.
Collapse
Affiliation(s)
- Devin I Alewel
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Urmila P Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
6
|
Herzberg MP, Nielsen AN, Luby J, Sylvester CM. Measuring neuroplasticity in human development: the potential to inform the type and timing of mental health interventions. Neuropsychopharmacology 2024; 50:124-136. [PMID: 39103496 PMCID: PMC11525577 DOI: 10.1038/s41386-024-01947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 08/07/2024]
Abstract
Neuroplasticity during sensitive periods, the molecular and cellular process of enduring neural change in response to external stimuli during windows of high environmental sensitivity, is crucial for adaptation to expected environments and has implications for psychiatry. Animal research has characterized the developmental sequence and neurobiological mechanisms that govern neuroplasticity, yet gaps in our ability to measure neuroplasticity in humans limit the clinical translation of these principles. Here, we present a roadmap for the development and validation of neuroimaging and electrophysiology measures that index neuroplasticity to begin to address these gaps. We argue that validation of measures to track neuroplasticity in humans will elucidate the etiology of mental illness and inform the type and timing of mental health interventions to optimize effectiveness. We outline criteria for evaluating putative neuroimaging measures of plasticity in humans including links to neurobiological mechanisms shown to govern plasticity in animal models, developmental change that reflects heightened early life plasticity, and prediction of neural and/or behavior change. These criteria are applied to three putative measures of neuroplasticity using electroencephalography (gamma oscillations, aperiodic exponent of power/frequency) or functional magnetic resonance imaging (amplitude of low frequency fluctuations). We discuss the use of these markers in psychiatry, envision future uses for clinical and developmental translation, and suggest steps to address the limitations of the current putative neuroimaging measures of plasticity. With additional work, we expect these markers will significantly impact mental health and be used to characterize mechanisms, devise new interventions, and optimize developmental trajectories to reduce psychopathology risk.
Collapse
Affiliation(s)
- Max P Herzberg
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA.
| | - Ashley N Nielsen
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA.
| | - Joan Luby
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Chad M Sylvester
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
7
|
Runyun ŞL, van Wassenhove V, Balci F. Altered temporal awareness during Covid-19 pandemic. PSYCHOLOGICAL RESEARCH 2024; 88:2335-2345. [PMID: 39034344 DOI: 10.1007/s00426-024-02004-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/05/2024] [Indexed: 07/23/2024]
Abstract
Social isolation during the COVID-19 pandemic had profound effects on human well-being. A handful of studies have focused on how time perception was altered during the COVID-19 pandemic, while no study has tested whether temporal metacognition is also affected by the lockdown. We examined the impact of long-term social isolation during the COVID-19 pandemic on the ability to monitor errors in timing performance. We recruited 1232 participants from 12 countries during lockdown, 211 of which were retested "post-pandemic" for within-group comparisons. We also tested a new group of 331 participants during the "post-pandemic" period and compared their data to those of 1232 participants tested during the lockdown (between-group comparison). Participants produced a 3600 ms target interval and assessed the magnitude and direction of their time production error. Both within and between-group comparisons showed reduced metric error monitoring performance during the lockdown, even after controlling for government-imposed stringency indices. A higher level of reported social isolation also predicted reduced temporal error monitoring ability. Participants produced longer duration during lockdown compared to post-lockdown (again controlling for government stringency indices). We reason that these effects may be underlain by altered biological and behavioral rhythms during social isolation experienced during the COVID-19 pandemic. Understanding these effects is crucial for a more complete characterization of the cognitive consequences of long-term social isolation.
Collapse
Affiliation(s)
- Şerife Leman Runyun
- Department of Psychology, Koç University, Istanbul, Turkey
- Department of Psychology, Northeastern University, 360 Huntington Ave, 125 NI, Boston, MA, 02115, USA
| | - Virginie van Wassenhove
- Cognitive Neuroimaging Unit, CEA, INSERM, CNRS, Université Paris-Saclay, NeuroSpin, Gif/Yvette, 91191, France
| | - Fuat Balci
- Department of Psychology, Koç University, Istanbul, Turkey.
- Department of Biological Sciences, University of Manitoba, 50 Sifton Road, Winnipeg, MB, R3T 2M5, Canada.
| |
Collapse
|
8
|
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress / adversity: Structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress 2024; 33:100669. [PMID: 39309367 PMCID: PMC11415888 DOI: 10.1016/j.ynstr.2024.100669] [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: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Adverse early life experiences are strongly associated with reduced cognitive function throughout life. The link is strong in many human studies, but these do not enable assigning causality, and the limited access to the live human brain can impede establishing the mechanisms by which early-life adversity (ELA) may induce cognitive problems. In experimental models, artificially imposed chronic ELA/stress results in deficits in hippocampus dependent memory as well as increased vulnerability to the deleterious effects of adult stress on memory. This causal relation of ELA and life-long memory impairments provides a framework to probe the mechanisms by which ELA may lead to human cognitive problems. Here we focus on the consequences of a one-week exposure to adversity during early postnatal life in the rodent, the spectrum of the ensuing memory deficits, and the mechanisms responsible. We highlight molecular, cellular and circuit mechanisms using convergent trans-disciplinary approaches aiming to enable translation of the discoveries in experimental models to the clinic.
Collapse
Affiliation(s)
- 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
| | - Matthew T. Birnie
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
9
|
Nipper MA, Helms ML, Finn DA, Ryabinin AE. Stress-enhanced ethanol drinking does not increase sensitivity to the effects of a CRF-R1 antagonist on ethanol intake in male and female mice. Alcohol 2024; 120:73-83. [PMID: 38185336 PMCID: PMC11326135 DOI: 10.1016/j.alcohol.2024.01.001] [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: 10/27/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 01/09/2024]
Abstract
Research confirms that stress is associated with alcohol drinking and relapse in males and females and that there are sex differences in the alcohol-related adaptations of stress pathways. The predator stress (PS) model of traumatic stress produces an increase in alcohol drinking or self-administration in a subpopulation of rodents, so it is utilized as an animal model of comorbid alcohol use disorder (AUD) and post-traumatic stress disorder (PTSD). Previous work determined that sensitivity to PS-enhanced drinking produced sex differences in proteins related to stress-regulating systems in the medial prefrontal cortex and hippocampus. The present studies examined whether male and female C57BL/6J mice differ in sensitivity to the ability of the corticotropin releasing factor receptor 1 antagonist CP-376395 to decrease PS-enhanced drinking. In control studies, CP-376395 doses of 5, 10, and 20 mg/kg dose-dependently decreased 4-h ethanol drinking. Next, CP-376395 doses of 5 and 10 mg/kg were tested for effects on ethanol drinking in mice with differential sensitivity to PS-enhanced drinking. Subgroups of "Sensitive" and "Resilient" male and female mice were identified based on changes in ethanol intake in an unrestricted-access ethanol-drinking procedure following four exposures to PS (dirty rat bedding). During the first 2 h post-injection of CP-376395, both doses significantly decreased ethanol licks versus vehicle in the females, with no significant interaction between subgroups, whereas the 10 mg/kg dose significantly decreased ethanol licks versus vehicle in the "Resilient" males. Thus, sensitivity to the suppressive effect of CP-376395 on stress-induced ethanol intake was greater in females versus males, whereas sensitivity and resilience to PS-enhanced drinking produced differential sensitivity to the ability of CP-376395 to decrease ethanol drinking only in male mice. Our results argue against greater efficacy of CRF-R1's ability to decrease ethanol intake in subjects with traumatic stress-enhanced ethanol drinking.
Collapse
Affiliation(s)
- Michelle A Nipper
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States
| | - Melinda L Helms
- Department of Research, VA Portland Health Care System, Portland, OR 97239, United States
| | - Deborah A Finn
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States; Department of Research, VA Portland Health Care System, Portland, OR 97239, United States
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States.
| |
Collapse
|
10
|
Hicks EM, Seah C, Deans M, Lee S, Johnston KJA, Cote A, Ciarcia J, Chakka A, Collier L, Holtzheimer PE, Young KA, Krystal JH, Brennand KJ, Nestler EJ, Girgenti MJ, Huckins LM. Decoding the transcriptomic signatures of psychological trauma in human cortex and amygdala. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619681. [PMID: 39484441 PMCID: PMC11526900 DOI: 10.1101/2024.10.23.619681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Psychological trauma has profound effects on brain function and precipitates psychiatric disorders in vulnerable individuals, however, the molecular mechanisms linking trauma with psychiatric risk remain incompletely understood. Using RNA-seq data postmortem brain tissue of a cohort of 304 donors (N=136 with trauma exposure), we investigated transcriptional signatures of trauma exposures in two cortical regions (dorsolateral prefrontal cortex, and dorsal anterior cingulate cortex) and two amygdala regions (medial amygdala and basolateral amygdala) associated with stress processing and regulation. We focused on dissecting heterogeneity of traumatic experiences in these transcriptional signatures by investigating exposure to several trauma types (childhood, adulthood, complex, single acute, combat, and interpersonal traumas) and interactions with sex. Overall, amygdala regions were more vulnerable to childhood traumas, whereas cortical regions were more vulnerable to adulthood trauma (regardless of childhood experience). Using cell-type-specific expression imputation, we identified a strong transcriptional response of medial amygdala excitatory neurons to childhood trauma, which coincided with dysregulation observed in a human induced pluripotent stem cell (hiPSC)-derived glutamatergic neurons exposed to hydrocortisone. We resolved multiscale coexpression networks for each brain region and identified modules enriched in trauma signatures and whose connectivity was altered with trauma. Trauma-associated coexpression modules provide insight into coordinated functional dysregulation with different traumas and point to potential gene targets for further dissection. Together, these data provide a characterization of the long-lasting human encoding of traumatic experiences in corticolimbic regions of human brain.
Collapse
|
11
|
Rong J, Wang Y, Liu N, Shen L, Ma Q, Wang M, Han B. Chronic stress induces insulin resistance and enhances cognitive impairment in AD. Brain Res Bull 2024; 217:111083. [PMID: 39304000 DOI: 10.1016/j.brainresbull.2024.111083] [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: 07/15/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Chronic stress can induce the cognitive impairment, and even promote the occurrence and development of Alzheimer's disease (AD). Evidence has suggested that chronic stress impacts on glucose metabolism, and both of these have been implicated in AD. Here we focused on the effect of insulin resistance in glucose metabolism, and further evaluated the changes in cognition and pathology. METHODS Male 9-month-old wild-type and APP/PS1 mice were randomly divided into 4 groups. Mice in the chronic unpredictable mild stress (CUMS) groups were exposed for 4 weeks. Homeostatic Model Assessment (HOMA) was utilized to evaluate insulin sensitivity. A total of eighty-four genes related to the insulin signaling pathway were examined for rapid screening. Additionally, the phosphorylated protein expressions of insulin receptors (IR), IR substrate 1 (IRS1), c-Jun N-terminal kinase (JNK), and amyloid were detected in the hippocampus. Cognitive function was assessed through ethological methods. Cognitive function was assessed using both the Morris water maze (MWM) and the Passive avoidance test (PAT). RESULTS Four weeks of CUMS exposure significantly increased the HOMA value, indicating reduced insulin sensitivity. The gene expressions of Insr and Lipe were downregulated. Additionally, the analysis revealed a significant interaction between the genotype (wild-type vs. APP/PS1) and CUMS treatment on the phosphorylated protein expressions of insulin receptor substrate 1 (IRS1). Specifically, CUMS exposure increased the inhibitory phosphorylation site (IRS1-pSer636) and decreased the excitatory phosphorylation site (IRS1-pTyr465) in the post-insulin receptor signaling pathway within the hippocampus of both wild-type and APP/PS1 mice. Moreover, CUMS exposure induced and exacerbated cognitive impairments in both wild-type and APP/PS1 mice, as assessed by the Morris water maze (MWM) and Passive avoidance test (PAT). However, there was no significant effect of CUMS on senile plaque deposition or levels of Aβ42 and Aβ40 in wild-type mice. CONCLUSIONS Chronic stress significantly affects hippocampal cognitive function through insulin resistance and exacerbates AD pathology. This study reveals the complex relationship between chronic stress, insulin resistance, and AD, providing new insights for developing interventions targeting chronic stress and insulin resistance.
Collapse
Affiliation(s)
- Jiaying Rong
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, Shijiazhuang, Hebei, China; Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Yanyong Wang
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, Shijiazhuang, Hebei, China; Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Na Liu
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, Shijiazhuang, Hebei, China; Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Li Shen
- Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qinying Ma
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, Shijiazhuang, Hebei, China; Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Mingwei Wang
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Bing Han
- Department of Neurology, Hebei Hospital, Xuanwu Hospital of Capital Medical University, Shijiazhuang, Hebei, China; Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Laboratory of Hebei Province, Shijiazhuang, Hebei, China.
| |
Collapse
|
12
|
Yi W, Chen W, Lan B, Yan L, Hu X, Wu J. A U-shaped relationship between chronic academic stress and the dynamics of reward processing. Neuroimage 2024; 300:120849. [PMID: 39265955 DOI: 10.1016/j.neuroimage.2024.120849] [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: 03/27/2024] [Revised: 08/29/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024] Open
Abstract
Despite the potential link between stress-induced reward dysfunctions and the development of mental problems, limited human research has investigated the specific impacts of chronic stress on the dynamics of reward processing. Here we aimed to investigate the relationship between chronic academic stress and the dynamics of reward processing (i.e., reward anticipation and reward consumption) using event-related potential (ERP) technology. Ninety healthy undergraduates who were preparing for the National Postgraduate Entrance Examination (NPEE) participated in the study and completed a two-door reward task, their chronic stress levels were assessed via the Perceived Stress Scale (PSS). The results showed that a lower magnitude of reward elicited more negative amplitudes of cue-N2 during the anticipatory phase, and reward omission elicited more negative amplitudes of FRN compared to reward delivery especially in high reward conditions during the consummatory phase. More importantly, the PSS score exhibited a U-shaped relationship with cue-N2 amplitudes regardless of reward magnitude during the anticipatory phase; and FRN amplitudes toward reward omission in high reward condition during the consummatory phase. These findings suggest that individuals exposed to either low or high levels of chronic stress, as opposed to moderate stress levels, exhibited a heightened reward anticipation, and an augmented violation of expectations or affective response when faced with relatively more negative outcomes.
Collapse
Affiliation(s)
- Wei Yi
- School of Psychology, Shenzhen University, 3688#, Nanhai Avenue, Nanshan District, Shenzhen 518060, China
| | - Wangxiao Chen
- School of Psychology, Shenzhen University, 3688#, Nanhai Avenue, Nanshan District, Shenzhen 518060, China
| | - Biqi Lan
- School of Psychology, Shenzhen University, 3688#, Nanhai Avenue, Nanshan District, Shenzhen 518060, China
| | - Linlin Yan
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Xiaoqing Hu
- Department of Psychology, The University of Hong Kong, Room 6.62, Jocky Club Tower, Pokfulam, Hong Kong, China
| | - Jianhui Wu
- School of Psychology, Shenzhen University, 3688#, Nanhai Avenue, Nanshan District, Shenzhen 518060, China.
| |
Collapse
|
13
|
Lindquist KA, Mecklenburg J, Hovhannisyan AH, Ruparel S, Akopian AN. Investigating Mechanically Activated Currents from Trigeminal Neurons of Non-Human Primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.06.616876. [PMID: 39416195 PMCID: PMC11482751 DOI: 10.1101/2024.10.06.616876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Introduction Pain sensation has predominantly mechanical modalities in many pain conditions. Mechanically activated (MA) ion channels on sensory neurons underly responsiveness to mechanical stimuli. The study aimed to address gaps in knowledge regarding MA current properties in higher order species such as non-human primates (NHP; common marmosets), and characterization of MA currents in trigeminal (TG) neuronal subtypes. Methods We employed patch clamp electrophysiology and immunohistochemistry (IHC) to associate MA current types to different marmoset TG neuronal groups. TG neurons were grouped according to presumed marker expression, action potential (AP) width, characteristic AP features, after-hyperpolarization parameters, presence/absence of AP trains and transient outward currents, and responses to mechanical stimuli. Results Marmoset TG were clustered into 5 C-fiber and 5 A-fiber neuronal groups. The C1 group likely represent non-peptidergic C-nociceptors, the C2-C4 groups resembles peptidergic C-nociceptors, while the C5 group could be either cold-nociceptors or C-low-threshold-mechanoreceptors (C-LTMR). Among C-fiber neurons only C4 were mechanically responsive. The A1 and A2 groups are likely A-nociceptors, while the A3-A5 groups probably denote different subtypes of A-low-threshold-mechanoreceptors (A-LTMRs). Among A-fiber neurons only A1 was mechanically unresponsive. IHC data was correlated with electrophysiology results and estimates that NHP TG has ∼25% peptidergic C-nociceptors, ∼20% non-peptidergic C-nociceptors, ∼30% A-nociceptors, ∼5% C-LTMR, and ∼20% A-LTMR. Conclusion Overall, marmoset TG neuronal subtypes and their associated MA currents have common and unique properties compared to previously reported data. Findings from this study could be the basis for investigation on MA current sensitizations and mechanical hypersensitivity during head and neck pain conditions.
Collapse
|
14
|
Liu L, Li R, Wu L, Guan Y, Miao M, Wang Y, Li C, Wu C, Lu G, Hu X, Sun L. (2R,6R)-hydroxynorketamine alleviates PTSD-like endophenotypes by regulating the PI3K/AKT signaling pathway in rats. Pharmacol Biochem Behav 2024; 245:173891. [PMID: 39369910 DOI: 10.1016/j.pbb.2024.173891] [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: 07/30/2024] [Revised: 09/08/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Patients diagnosed with post-traumatic stress disorder (PTSD) mainly exhibit enduring adverse emotions, heightening susceptibility to suicidal thoughts and behaviors. Notably, metabolites of ketamine, particularly (2R,6R)-hydroxyketamine (HNK), have demonstrated favorable antidepressant properties. However, the precise mechanism through which HNK exerts its therapeutic effects on negative emotional symptoms in PTSD patients should be fully elucidated. METHODS In this investigation, a model involving a single prolonged stress and plantar shock (SPS&S) was utilized, followed by the administration of (2R, 6R)-HNK into the lateral ventricle subsequent to the recovery phase. The evaluation of PTSD-related behaviors was conducted through the open field test (OFT), elevated plus maze test (EMPT), and forced swim test (FST). The expression of phosphatidylinositol 3-kinase (PI3K)/phosphokinase B (AKT) signaling pathway in rat brain regions was analyzed using molecular biology experiments. RESULTS SPS&S rats displayed adverse emotional behaviors characterized by depression and anxiety. Treatment with (2R, 6R)-HNK enhanced exploratory behavior and reversed negative emotional behaviors. This intervention mitigated disruptions in the expression levels of PI3K/AKT signaling pathway-associated proteins in the HIP and PFC, without influencing PI3K/AKT signaling in the AMY of SPS&S rats. CONCLUSION Traumatic stress can trigger negative emotional reactions in rats, potentially involving the PI3K/AKT signaling pathway in the HIP, PFC, and AMY. The (2R, 6R)-HNK compounds have demonstrated the potential to mitigate adverse emotions in rats subjected to the SPS&S paradigm. This effect may be attributed to the modulation of the PI3K/AKT signaling pathway in the HIP, and PFC, with a particularly notable impact observed in the HIP region.
Collapse
Affiliation(s)
- Lifen Liu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Rui Li
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Lanxia Wu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yubo Guan
- School of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Miao Miao
- School of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yuxuan Wang
- School of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Changjiang Li
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Chunyan Wu
- Department of Neurology, Affiliated Hospital of Shandong Second Medical University, Weifang, PR China
| | - Guohua Lu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Xinyu Hu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China; CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, PR China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, PR China.
| | - Lin Sun
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China; Department of Neurosurgery, Shanting District People's Hospital, Beijing Road, New Town, Zaozhuang, Shandong 277200, PR China; Management Committee of Shanting Economic Development Zone, No.37, Fuqian Road, Zaozhuang, Shandong 277200, PR China.
| |
Collapse
|
15
|
Zheng Z, Liu Y, Mu R, Guo X, Feng Y, Guo C, Yang L, Qiu W, Zhang Q, Yang W, Dong Z, Qiu S, Dong Y, Cui Y. A small population of stress-responsive neurons in the hypothalamus-habenula circuit mediates development of depression-like behavior in mice. Neuron 2024:S0896-6273(24)00660-3. [PMID: 39389052 DOI: 10.1016/j.neuron.2024.09.012] [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: 12/11/2023] [Revised: 06/25/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Accumulating evidence has shown that various brain functions are associated with experience-activated neuronal ensembles. However, whether such neuronal ensembles are engaged in the pathogenesis of stress-induced depression remains elusive. Utilizing activity-dependent viral strategies in mice, we identified a small population of stress-responsive neurons, primarily located in the middle part of the lateral hypothalamus (mLH) and the medial part of the lateral habenula (LHbM). These neurons serve as "starter cells" to transmit stress-related information and mediate the development of depression-like behaviors during chronic stress. Starter cells in the mLH and LHbM form dominant connections, which are selectively potentiated by chronic stress. Silencing these connections during chronic stress prevents the development of depression-like behaviors, whereas activating these connections directly elicits depression-like behaviors without stress experience. Collectively, our findings dissect a core functional unit within the LH-LHb circuit that mediates the development of depression-like behaviors in mice.
Collapse
Affiliation(s)
- Zhiwei Zheng
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yiqin Liu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Ruiqi Mu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Xiaonan Guo
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yirong Feng
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Chen Guo
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Liang Yang
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Wenxi Qiu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Qi Zhang
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhaoqi Dong
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shuang Qiu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yiyan Dong
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China.
| | - Yihui Cui
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
16
|
Lugenbühl JF, Viho EMG, Binder EB, Daskalakis NP. Stress molecular signaling in interaction with cognition. Biol Psychiatry 2024:S0006-3223(24)01628-7. [PMID: 39368530 DOI: 10.1016/j.biopsych.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/02/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
Exposure to stressful life events is associated with a high risk of developing psychiatric disorders with a wide variety of symptoms. Cognitive symptoms in stress-related psychiatric disorders can be particularly challenging to understand, both for those experiencing them and for healthcare providers. To gain insights, it is important to capture stress-induced structural, epigenomic, transcriptomic, and proteomic changes in relevant brain regions such as the amygdala, hippocampus, locus coeruleus and prefrontal cortex, resulting in long-lasting alterations in brain function. In this review, we will emphasize a subset of stress molecular mechanisms altering neuroplasticity, neurogenesis, and balance between excitatory and inhibitory neurons. We then discuss how to identify genetic risk factors that may accelerate stress-driven or stress-induced cognitive impairment. Despite the development of new technologies such as single-cell resolution sequencing, our understanding of the molecular effects of stress in the brain remains to be deepened. A better understanding of the diversity of stress effects in different brain regions and cell types is a pre-requisite to open new avenues for mechanism-informed prevention and treatment of stress-related cognitive symptoms.
Collapse
Affiliation(s)
- Justina F Lugenbühl
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry and Neuropsychology, School for Mental Health, and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
| | - Eva M G Viho
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
| | - Nikolaos P Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
17
|
Pawluski JL. The parental brain, perinatal mental illness, and treatment: A review of key structural and functional changes. Semin Perinatol 2024; 48:151951. [PMID: 39030131 DOI: 10.1016/j.semperi.2024.151951] [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] [Indexed: 07/21/2024]
Abstract
The transition to parenthood is perhaps the only time in adult life when the brain changes to such a significant degree in such a short period, particularly in birthing parents. It is also a time when there is an increased risk of developing a mental illness, which may be due, in part, to the increased neuroplasticity. Thus, we must develop interventions and treatments that support parents and promote parental brain health. This review will highlight key findings from current research on how human brain structure and function are modified with 1) the transition to parenthood, 2) parenting stress and perinatal mental illness, and 3) treatments aimed at promoting perinatal mental health. The focus will be on birthing parents and mothers, but brain changes in non-birthing parents will also be discussed. Improvements in our understanding of the parental brain, in health and with illness, will promote the well-being of generations to come.
Collapse
Affiliation(s)
- Jodi L Pawluski
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
18
|
Trivedi M, Dumkrieger G, Chong CD, Leibovit-Reiben Z, Schwedt TJ. A history of abuse is associated with more severe migraine- and pain-related disability: Results from the American Registry for Migraine Research. Headache 2024; 64:1109-1123. [PMID: 39051483 DOI: 10.1111/head.14787] [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: 10/12/2023] [Revised: 06/06/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Prior studies have established an association between a history of abuse and more severe migraine presentation. OBJECTIVES This cross-sectional, observational study of a clinic-based migraine population used validated measures to elucidate migraine-specific and migraine-related burdens among patients with a history of abuse. METHODS Patients with migraine (n = 866) from the American Registry for Migraine Research self-reported if they had a history of emotional, physical, and/or sexual abuse and completed questionnaires assessing migraine-related burden: Migraine Disability Assessment, Subjective Cognitive Impairment Scale for Migraine Attacks, Work Productivity and Activity Impairment, Patient-Reported Outcomes Measurement Information System Pain Interference, Patient Health Questionnaire-2, and Generalized Anxiety Disorder-7. Migraine-related burden in patients with versus without a history of abuse was compared. Subsequently, a mediation analysis evaluated the impact of depression and anxiety symptoms in the relationship between abuse history and migraine burden. RESULTS A history of abuse was reported by 36.5% (n = 316/866) of participants. After controlling for patient age, sex, years lived with headache, and headache frequency, a history of abuse was significantly associated with more severe migraine-related disability. The combined burden of depression and anxiety symptoms mediated the relationship. CONCLUSION A history of abuse is associated with greater migraine-related disability. Future studies should determine if identification and management of the psychological and physical sequelae of abuse reduce migraine burden.
Collapse
Affiliation(s)
- Meesha Trivedi
- Department of Neurology, Mayo Clinic, Phoenix, Arizona, USA
- Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | | | | | - Zachary Leibovit-Reiben
- Department of Neurology, Mayo Clinic, Phoenix, Arizona, USA
- College of Medicine - Tucson, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Todd J Schwedt
- Department of Neurology, Mayo Clinic, Phoenix, Arizona, USA
| |
Collapse
|
19
|
Liu X, Liu R, Sun YX, Wang HL, Wang H, Wang T, Ma YN, Li XX, Wang Q, Su YA, Li JT, Si TM. Dorsal CA3 overactivation mediates witnessing stress-induced recognition memory deficits in adolescent male mice. Neuropsychopharmacology 2024; 49:1666-1677. [PMID: 38504012 PMCID: PMC11399124 DOI: 10.1038/s41386-024-01848-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/18/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
Witnessing violent or traumatic events is common during childhood and adolescence and could cause detrimental effects such as increased risks of psychiatric disorders. This stressor could be modeled in adolescent laboratory animals using the chronic witnessing social defeat (CWSD) paradigm, but the behavioral consequences of CWSD in adolescent animals remain to be validated for cognitive, anxiety-like, and depression-like behaviors and, more importantly, the underlying neural mechanisms remain to be uncovered. In this study, we first established the CWSD model in adolescent male mice and found that CWSD impaired cognitive function and increased anxiety levels and that these behavioral deficits persisted into adulthood. Based on the dorsal-ventral functional division in hippocampus, we employed immediate early gene c-fos immunostaining after behavioral tasks and found that CWSD-induced cognition deficits were associated with dorsal CA3 overactivation and anxiety-like behaviors were associated with ventral CA3 activity reduction. Indeed, chemogenetic activation and inhibition of dorsal CA3 neurons mimicked and reversed CWSD-induced recognition memory deficits (not anxiety-like behaviors), respectively, whereas both inhibition and activation of ventral CA3 neurons increased anxiety-like behaviors in adolescent mice. Finally, chronic administration of vortioxetine (a novel multimodal antidepressant) successfully restored the overactivation of dorsal CA3 neurons and the cognitive deficits in CWSD mice. Together, our findings suggest that dorsal CA3 overactivation mediates CWSD-induced recognition memory deficits in adolescent male mice, shedding light on the pathophysiology of adolescent CWSD-induced adverse effects and providing preclinical evidence for early treatment of stress-induced cognitive deficits.
Collapse
Affiliation(s)
- Xiao Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Rui Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ya-Xin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Hong-Li Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Han Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ting Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Yu-Nu Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xue-Xin Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Qi Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
| |
Collapse
|
20
|
Klausing AD, Fukuwatari T, DeAngeli N, Bucci DJ, Schwarcz R. Adrenalectomy exacerbates stress-induced impairment in fear discrimination: A causal role for kynurenic acid? Biochem Pharmacol 2024; 228:116350. [PMID: 38852644 DOI: 10.1016/j.bcp.2024.116350] [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: 04/10/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Impaired activity of the hypothalamic-pituitary axis and reduced blood levels of glucocorticoids (GCs) are signature features of stress-related maladies. Recent evidence suggests a possible role of the tryptophan metabolite kynurenic acid (KYNA) in this context. Here we investigated possible causal relationships in adult male rats, using stress-induced fear discrimination as a translationally relevant behavioral outcome measure. One week following adrenalectomy (ADX) or sham surgery, animals were for 2 h either physically restrained or exposed to a predator odor, which caused a much milder stress response. Extracellular KYNA levels were determined before, during and after stress by in vivo microdialysis in the prefrontal cortex. Separate cohorts underwent a fear discrimination procedure starting immediately after stress termination. Different auditory conditioned stimuli (CS) were either paired with a foot shock (CS+) or non-reinforced (CS-). One week later, fear was assessed by re-exposing the animals to each CS. Separate groups of rats were treated with the KYNA synthesis inhibitor BFF-816 prior to stress initiation to test a causal role of KYNA in fear discrimination. Restraint stress raised extracellular KYNA levels by ∼85 % in ADX rats for several hours, and these animals were unable to discriminate between CS+ and CS-. Both effects were prevented by BFF-816 and were not observed after exposure to predator odor or in sham-operated rats. These findings suggest that a causal connection exists between adrenal function, stress-induced KYNA increases, and behavioral deficits. Pharmacological inhibition of KYNA synthesis may therefore be an attractive, novel option for the treatment of stress-related disorders.
Collapse
Affiliation(s)
- Alex D Klausing
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tsutomu Fukuwatari
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nicole DeAngeli
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - David J Bucci
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Zhao J, Xue E, Zhou S, Zhang M, Jiang F, Wu W, Liu Z, Li X. Allostatic load increases the incidence and risk of adverse prognosis in inflammatory bowel disease. Aliment Pharmacol Ther 2024; 60:1062-1074. [PMID: 39166843 DOI: 10.1111/apt.18217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/31/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Elevated allostatic load (AL) has been associated with the risk and poor prognosis of many chronic diseases. The association between AL and inflammatory bowel disease (IBD) is unknown. AIMS The aim of this study is to investigate the associations between AL and the risk and prognosis of IBD. METHODS We included 326,345 adults and 3767 patients with IBD from the UK Biobank. AL served as the exposure, estimated using the AL biomarker panel, with the primary outcomes including the risk and prognosis of IBD. We used Cox regression models to examine the associations. RESULTS High AL biomarker panel was associated with a greater risk of IBD (hazard ratio: 1.19, 95% CI: 1.08-1.31), ulcerative colitis (1.17, 95%CI: 1.04-1.32), and Crohn's disease (1.25, 95%CI: 1.05-1.49). Risk of developing IBD increased by 12% in quartile 2, 20% in quartile 3, and 37% in quartile 4 as AL biomarker panel increased. The all-cause mortality risk in IBD compared with quartile 1 rose by 54% for quartile 2, 72% for quartile 3, and 82% for quartile 4, as AL biomarker panel increased. Similar effects were also observed for ulcerative colitis and Crohn's disease. An increase in AL biomarker panel count was associated with an elevated risk of intestinal resection and colorectal cancer in IBD. CONCLUSIONS Increased AL is associated with IBD risk, as well as the risks of intestinal resection, colorectal cancer and mortality.
Collapse
Affiliation(s)
- Jianhui Zhao
- Department of Big Data in Health Science, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Erxu Xue
- Nursing Department, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Siyun Zhou
- Department of Big Data in Health Science, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Meng Zhang
- Department of Big Data in Health Science, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fangyuan Jiang
- Department of Big Data in Health Science, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei Wu
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, the Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhanju Liu
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, the Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xue Li
- Department of Big Data in Health Science, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Xing S, Xu S, Wang L, Guo L, Zhou X, Wu H, Wang W, Liu L. Salidroside exerts antidepressant-like action by promoting adult hippocampal neurogenesis through SIRT1/PGC-1α signalling. Acta Neuropsychiatr 2024:1-11. [PMID: 39344773 DOI: 10.1017/neu.2024.28] [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] [Indexed: 10/01/2024]
Abstract
Depression is one of the major mental disorders, which seriously endangers human health, brings a serious burden to patients’ families. In this study, we intended to further explore the antidepressant-like effect and possible molecular mechanisms of Salidroside (SAL). We built corticosterone (CORT)-induced depressive mice model and used behavioural tests to evaluate depression behaviour. To explore the molecular mechanisms of SAL, we employed a variety of methods such as immunofluorescence, western blot, pharmacological interference, etc. The results demonstrated that SAL both at 25 mg/kg and 50 mg/kg can reduce immobility time in the tail suspension test (TST). At the same time, SAL treatment could restore the reduced sugar water intake preference in the sucrose preference test (SPT) in CORT-induced depressive mice and reduce the immobility time in TST and forced swimming experiments (FST). In addition, SAL treatment reversed the reduction in the number of Ki-67, BrdU, and NeuN in the hippocampus due to CORT treatment. SAL treatment also restored the expression of SIRT1, PGC-1α, brain-derived neurotrophic factor (BDNF) and other proteins in the hippocampus. In addition, after blocking SIRT1 signalling with EX527, we found that the treatment with SAL failed to reduce the immobility time in TST and FST, the level of SIRT1 and PGC-1α activity were correspondingly downregulated, and the expression of DCX and Ki-67 in the hippocampus failed to be activated. These findings suggested that SAL exerts antidepressant-like effects by promoting hippocampal neurogenesis through the SIRT1/PGC-1α signalling pathway.
Collapse
Affiliation(s)
- Shan Xing
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuyi Xu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Linjiao Wang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyuan Guo
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haoxin Wu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lanying Liu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai, China
- Department of Psychiatry, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Mental Diseases of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
23
|
Svob Strac D. Genetic Basis of Stress-Related Neuropsychiatric Disorders. Genes (Basel) 2024; 15:1274. [PMID: 39457398 PMCID: PMC11507372 DOI: 10.3390/genes15101274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Stress exposure is one of the major risk factors for the development of different psychiatric and neurodegenerative diseases, leading to various (mal)adaptive alternations in different parts of the brain, and affecting both brain structure and function [...].
Collapse
Affiliation(s)
- Dubravka Svob Strac
- Rudjer Boskovic Institute, Division of Molecular Medicine, Bijenicka Cesta 54, 10000 Zagreb, Croatia
| |
Collapse
|
24
|
Gobbi C, Sánchez-Marín L, Flores-López M, Medina-Vera D, Pavón-Morón FJ, Rodríguez de Fonseca F, Serrano A. Sex-dependent effects of acute stress and alcohol exposure during adolescence on mRNA expression of brain signaling systems involved in reward and stress responses in young adult rats. Biol Sex Differ 2024; 15:75. [PMID: 39327618 PMCID: PMC11426001 DOI: 10.1186/s13293-024-00649-5] [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: 06/12/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Adolescent stress and alcohol exposure increase the risk of maladaptive behaviors and mental disorders in adulthood, with distinct sex-specific differences. Understanding the mechanisms underlying these early events is crucial for developing targeted prevention and treatment strategies. METHODS Male and female Wistar rats were exposed to acute restraint stress and intermittent alcohol during adolescence. We assessed lasting effects on plasma corticosterone (CORT) and adrenocorticotropic hormone (ACTH) levels, and mRNA expression of genes related to corticotropin releasing hormone (CRH), neuropeptide Y (NPY), corticoid, opioid, and arginine vasopressin systems in the amygdala and hypothalamus. RESULTS The main findings are as follows: (1) blood alcohol concentrations (BAC) increased after the final alcohol administration, but stressed males had lower BAC than non-stressed males; (2) Males gained significantly more weight than females; (3) Stressed females showed higher ACTH levels than non-stressed females, with no changes in males; (4) Stress increased CORT levels in males, while stressed, alcohol-treated females had lower CORT levels than non-stressed females; (5) CRH: Females had lower Crhr1 levels in the amygdala, while alcohol reduced Crhr2 levels in males but not females. Significant interactions among sex, stress, and alcohol were found in the hypothalamus, with distinct patterns between sexes; (6) NPY: In the amygdala, stress reduced Npy and Npy1r levels in males but increased them in females. Alcohol decreased Npy2r levels in males, with varied effects in females. Similar sex-specific patterns were observed in the hypothalamus; (7) Corticoid system: Stress and alcohol had complex, sex-dependent effects on Pomc, Nr3c1, and Nr3c2 in both brain regions; (8) Opioid receptors: Stress and alcohol blunted the elevated expression of Oprm1, Oprd1, and Oprk1 in the amygdala of males and the hypothalamus of females; (8) Vasopressin: Stress and alcohol interacted significantly to affect Avp and Avpr1a expression in the amygdala, with stronger effects in females. In the hypothalamus, alcohol increased Avp levels in females. CONCLUSIONS This study demonstrates that adolescent acute stress and alcohol exposure induce lasting, sex-specific alterations in systems involved in reward and stress responses. These findings emphasize the importance of considering sex differences in the prevention and management of HPA dysfunction and psychiatric disorders.
Collapse
Grants
- PI19/00886, PI20/01399, PI22/00427 and PI22/01833 Instituto de Salud Carlos III (ISCIII), Ministerio de Ciencia e Innovación and European Regional Development Funds-European Union (ERDF-EU)
- PI19/00886, PI20/01399, PI22/00427 and PI22/01833 Instituto de Salud Carlos III (ISCIII), Ministerio de Ciencia e Innovación and European Regional Development Funds-European Union (ERDF-EU)
- PI19/00886, PI20/01399, PI22/00427 and PI22/01833 Instituto de Salud Carlos III (ISCIII), Ministerio de Ciencia e Innovación and European Regional Development Funds-European Union (ERDF-EU)
- PT20-00101 Plataforma de biobanco y biomodelos animales y 3D de Málaga
- PT20-00101 Plataforma de biobanco y biomodelos animales y 3D de Málaga
- RD21/0009/0003 Programa RICORS RIAPAD
- RD21/0009/0003 Programa RICORS RIAPAD
- PNSD 2022/020 Ministerio de Sanidad, Delegación de Gobierno para el Plan Nacional sobre Drogas
Collapse
Affiliation(s)
- Carlotta Gobbi
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA- Plataforma BIONAND), Málaga, 29590, Spain
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga, 29010, Spain
| | - Laura Sánchez-Marín
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA- Plataforma BIONAND), Málaga, 29590, Spain
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga, 29010, Spain
| | - María Flores-López
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA- Plataforma BIONAND), Málaga, 29590, Spain
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga, 29010, Spain
| | - Dina Medina-Vera
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA- Plataforma BIONAND), Málaga, 29590, Spain
- Unidad Clínica Área del Corazón, Hospital Universitario Virgen de la Victoria de Málaga, Málaga, 29010, Spain
| | - Francisco Javier Pavón-Morón
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA- Plataforma BIONAND), Málaga, 29590, Spain.
- Unidad Clínica Área del Corazón, Hospital Universitario Virgen de la Victoria de Málaga, Málaga, 29010, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, 28029, Spain.
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA- Plataforma BIONAND), Málaga, 29590, Spain.
- Unidad de Gestión Clínica de Neurología, Hospital Regional Universitario de Málaga, Málaga, 29010, Spain.
| | - Antonia Serrano
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA- Plataforma BIONAND), Málaga, 29590, Spain
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga, 29010, Spain
| |
Collapse
|
25
|
Riopel C, Trudel X, Milot A, Laurin D, Gilbert-Ouimet M, Brisson C. Effort-reward imbalance at work assessed at midlife and prediabetes prevalence assessed 18 years later in a prospective cohort of white-collar workers. Ann Work Expo Health 2024:wxae071. [PMID: 39312482 DOI: 10.1093/annweh/wxae071] [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: 05/12/2023] [Accepted: 09/20/2024] [Indexed: 09/25/2024] Open
Abstract
OBJECTIVES Evidence suggests that workers exposed to psychosocial stressors at work from the effort-reward imbalance (ERI) model are at increased risk for type 2 diabetes mellitus (T2DM). However, evidence about the effect of ERI on prediabetes is scarce. This study aimed to examine the association between effort-reward imbalance at work, glycated hemoglobin level and the prevalence of prediabetes in women and men from a prospective cohort study. METHODS This study was conducted among 1354 white-collar workers followed for an average of 18 years. Effort-reward imbalance at work was measured in 1999 to 2001 using a validated instrument. Glycated hemoglobin was assessed at follow-up (2015 to 2018). Differences in mean glycated hemoglobin levels were estimated with linear models. Prediabetes prevalence ratios (PRs) were computed using robust Poisson regression models. RESULTS In women, those exposed to effort-reward imbalance at work at baseline had a higher prevalence of prediabetes (PR = 1.60, 95% confidence interval: 1.02-2.49) at follow-up following adjustment for sociodemographic, lifestyle-related, clinical, and other occupational risk factors. There was no difference in mean glycated hemoglobin levels. CONCLUSION Among women, effort-reward imbalance at work at midlife was associated with the prevalence of prediabetes, at older age. Preventive workplace interventions aiming to reduce the prevalence of effort-reward imbalance at work may be effective to reduce the prevalence of prediabetes among women.
Collapse
Affiliation(s)
- Camille Riopel
- Département de médecine sociale et préventive, Faculté de médecine, Université Laval, 1050 Av. de la Médecine, Québec, QC G1V 0A6, Canada
- Centre de recherche du CHU de Québec-Université Laval, 1050 Ch Ste-Foy, Québec, QC G1S 4L8, Canada
| | - Xavier Trudel
- Département de médecine sociale et préventive, Faculté de médecine, Université Laval, 1050 Av. de la Médecine, Québec, QC G1V 0A6, Canada
- Centre de recherche du CHU de Québec-Université Laval, 1050 Ch Ste-Foy, Québec, QC G1S 4L8, Canada
| | - Alain Milot
- Département de médecine sociale et préventive, Faculté de médecine, Université Laval, 1050 Av. de la Médecine, Québec, QC G1V 0A6, Canada
- Centre de recherche du CHU de Québec-Université Laval, 1050 Ch Ste-Foy, Québec, QC G1S 4L8, Canada
| | - Danielle Laurin
- Centre de recherche du CHU de Québec-Université Laval, 1050 Ch Ste-Foy, Québec, QC G1S 4L8, Canada
- Centre d'excellence sur le vieillissement de Québec, Centre de recherche VITAM en santé durable, CIUSSS-Capitale Nationale, 2480, chemin de la Canardière, Québec, QC G1J 2G1, Canada
- Faculty of Pharmacy, Université Laval, 1050 Av. de la Médecine, Québec, QC G1V 0A6, Canada
| | - Mahée Gilbert-Ouimet
- Centre de recherche du CHU de Québec-Université Laval, 1050 Ch Ste-Foy, Québec, QC G1S 4L8, Canada
- Health Sciences Department, Université du Québec à Rimouski Campus de Lévis, 1595 Bd Alphonse-Desjardins, Lévis, QC G6V 0A6, Canada
- Canada Research Chair in Sex and Gender in Occupational Health, Université du Québec à Rimouski Campus de Lévis, 1595 Bd Alphonse-Desjardins, Lévis, QC G6V 0A6, Canada
| | - Chantal Brisson
- Département de médecine sociale et préventive, Faculté de médecine, Université Laval, 1050 Av. de la Médecine, Québec, QC G1V 0A6, Canada
- Centre de recherche du CHU de Québec-Université Laval, 1050 Ch Ste-Foy, Québec, QC G1S 4L8, Canada
- Centre d'excellence sur le vieillissement de Québec, Centre de recherche VITAM en santé durable, CIUSSS-Capitale Nationale, 2480, chemin de la Canardière, Québec, QC G1J 2G1, Canada
| |
Collapse
|
26
|
Wu L, Zhang S, Zhang M, Ou Y, Chen J. Occlusal disharmony promotes anxiety-like behaviours by suppressing Sirt1. Clin Oral Investig 2024; 28:526. [PMID: 39276172 DOI: 10.1007/s00784-024-05918-6] [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] [Received: 05/27/2024] [Accepted: 09/08/2024] [Indexed: 09/16/2024]
Abstract
BACKGROUND Previous studies have indicated that occlusal disharmony (OD) can promote anxiety-like behaviours. However, the specific molecules involved in the development of anxiety-like behaviours and their underlying mechanisms remain unknown. METHODS OD was produced by anterior crossbite of female mice. We measured the anxiety levels of mice in each group and screened the hippocampal mRNA expression profiles of mice in the control group and OD group. The role of target mRNA in OD-induced anxiety-like behaviours was evaluated and we preliminarily explored the possible downstream pathways. RESULTS The results suggested that OD can induce and promote anxiety-like behaviours with/without chronic unpredictable mild stress. We found that Sirt1 was significantly downregulated within the hippocampus in OD mice. In addition, the downregulation of Sirt1 within the hippocampus in OD and control mice promoted anxiety-like behaviours, increased acetylated histone H3 expression and decreased Dnah12 transcription levels. In contrast, in OD mice subjected to an injection of resveratrol, there was a remission of anxiety-like behaviours and an upregulation of Sirt1 in the hippocampus, the effects of which were accompanied by decreased acetylated histone H3 expression and increased Dnah12 transcription levels. CONCLUSIONS OD leads to increased sensitivity to chronic stress in mice, resulting in anxiety-like behaviours. During this process, Sirt1 acts as an effective factor in the regulation of OD-induced anxiety-like behaviours. CLINICAL RELEVANCE OD, as a stressor, could induce anxiety-like behaviours. It investigates the impact of OD (a stressor) on the molecular genetic of the pathophysiology of major neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ling Wu
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Sihui Zhang
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Mi Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yanjing Ou
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jiang Chen
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Oral Diseases & Stomatological Key Lab of Fujian, School and Hospital of Stomatology, Fujian Medical University, 246 Yang Qiao Zhong Road, Fuzhou, China.
| |
Collapse
|
27
|
Ju Q, Gan Y, Peng H, Li B, Nie S, Schwarzer R. Does stress compromise fruit and vegetable intake? A randomized controlled trial testing a model with planning as a mediator and stress as a moderator. Nutrition 2024; 129:112581. [PMID: 39471754 DOI: 10.1016/j.nut.2024.112581] [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: 07/31/2023] [Revised: 07/10/2024] [Accepted: 09/03/2024] [Indexed: 11/01/2024]
Abstract
OBJECTIVE Individuals experiencing higher stress levels tend to consume fewer fruits and vegetables compared to their less stressed counterparts. Thus, to promote fruit and vegetable (FV) consumption, action planning has been proven effective in translating behavioral intentions into actual dietary behaviors. This study aims to evaluate a 7-day intervention designed to improve FV planning and intake, while also examining the role of stress. METHODS The trial employed a 3 (time: pretest, post-test, and follow-up) * 2 (group: intervention vs. control) between-participant factorial design. A total of 99 young Chinese adults (age = 23.84 years ± 4.63, 26 men) who had formed an explicit intention to consume more fruit and vegetables, participated in a 7-day online randomized controlled trial. RESULTS The intervention successfully enhanced FV planning as well as FV intake. Furthermore, a moderated mediation model revealed that FV planning mediated the relationship between experimental conditions and FV intake, with stress moderating this mediation. Specifically, planning facilitated FV intake for individuals with low stress levels, while this effect was not observed for those with high stress levels. CONCLUSION These findings confirm the positive impact of the planning intervention on improving FV intake, particularly for individuals with low stress levels, and highlight stress as a barrier to health behavior change that warrants further attention in future studies.
Collapse
Affiliation(s)
- Qianqian Ju
- School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental Health, Peking University, Beijing, China
| | - Yiqun Gan
- School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental Health, Peking University, Beijing, China.
| | - Huini Peng
- School of Psychological and Cognitive Sciences, Beijing Key Laboratory of Behavior and Mental Health, Peking University, Beijing, China
| | - Binghui Li
- Department of Psychology, National University of Singapore, Singapore, Singapore
| | - Shu Nie
- School of Public Administration, Inner Mongolia University, Inner Mongolia, China
| | - Ralf Schwarzer
- SWPS University, Wroclaw, Poland; Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
28
|
Matthews DB, Kerr E. Interaction of age and sex as factors in understanding the anxiolytic effects of alcohol: Unasked questions limiting the understanding of a critical health issue. Pharmacol Biochem Behav 2024; 245:173881. [PMID: 39278601 DOI: 10.1016/j.pbb.2024.173881] [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: 06/25/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Understanding the reasons why people consume alcohol is a critical health issue. Alcohol produces a variety of effects, including a reduction in stress or negative emotional states termed an anxiolytic effect. The anxiolytic effect of alcohol is an often-reported reason for why people begin consuming the drug. However, several factors concerning the stress-reducing effect of alcohol need to be investigated. For example, research has demonstrated that both age and sex are factors that impact alcohol's anxiolytic effect producing differential outcomes in aged female rats compared to aged male rats. In light of these findings, the current commentary highlights critical questions in need of research with the goal of better understanding how age and sex interact to influence the anxiolytic effect of alcohol. For example, the central nucleus of the amygdala has been identified as a critical brain region mediating the anxiolytic effect of drugs, but additional research is needed to understand how aging alters the neurological functioning of the central nucleus of the amygdala in both females and males. Furthermore, specific receptor isoforms, such as GABAA receptor α2, have been shown to be critical for anxiolysis and understanding how aging and sex alter receptor isoform expression by brain region is needed. Finally, age and sex interact to alter allopregnanolone levels in brain and differential neurosteroid levels may mediate alcohol's unique anxiolytic effect in aged female rats compared to aged male rats. Given the increasing age of the population in most countries and the increasing alcohol consumption levels in females compared to males, investigating the interaction of sex and age on alcohol's anxiolytic effect has great promise to discover critical answers to what are currently unasked questions.
Collapse
Affiliation(s)
- Douglas B Matthews
- Department of Psychology, University of Wisconsin - Eau Claire, Eau Claire, WI 54701, United States
| | - Emily Kerr
- Department of Psychology, University of Wisconsin - Eau Claire, Eau Claire, WI 54701, United States
| |
Collapse
|
29
|
Mo JW, Kong PL, Ding L, Fan J, Ren J, Lu CL, Guo F, Chen LY, Mo R, Zhong QL, Wen YL, Gu TT, Wang QW, Li SJ, Guo T, Gao TM, Cao X. Lysosomal TFEB-TRPML1 Axis in Astrocytes Modulates Depressive-like Behaviors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2403389. [PMID: 39264289 DOI: 10.1002/advs.202403389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/09/2024] [Indexed: 09/13/2024]
Abstract
Lysosomes are important cellular structures for human health as centers for recycling, signaling, metabolism and stress adaptation. However, the potential role of lysosomes in stress-related emotions has long been overlooked. Here, it is found that lysosomal morphology in astrocytes is altered in the medial prefrontal cortex (mPFC) of susceptible mice after chronic social defeat stress. A screen of lysosome-related genes revealed that the expression of the mucolipin 1 gene (Mcoln1; protein: mucolipin TRP channel 1) is decreased in susceptible mice and depressed patients. Astrocyte-specific knockout of mucolipin TRP channel 1 (TRPML1) induced depressive-like behaviors by inhibiting lysosomal exocytosis-mediated adenosine 5'-triphosphate (ATP) release. Furthermore, this stress response of astrocytic lysosomes is mediated by the transcription factor EB (TFEB), and overexpression of TRPML1 rescued depressive-like behaviors induced by astrocyte-specific knockout of TFEB. Collectively, these findings reveal a lysosomal stress-sensing signaling pathway contributing to the development of depression and identify the lysosome as a potential target organelle for antidepressants.
Collapse
Affiliation(s)
- Jia-Wen Mo
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Peng-Li Kong
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li Ding
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jun Fan
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jing Ren
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Cheng-Lin Lu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510260, China
| | - Fang Guo
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Liang-Yu Chen
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ran Mo
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qiu-Ling Zhong
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - You-Lu Wen
- Department of Psychology and Behavior, Guangdong 999 Brain Hospital, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510515, China
| | - Ting-Ting Gu
- Department of Psychology and Behavior, Guangdong 999 Brain Hospital, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510515, China
| | - Qian-Wen Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shu-Ji Li
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ting Guo
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tian-Ming Gao
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiong Cao
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510260, China
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
30
|
Arora I, Mal P, Arora P, Paul A, Kumar M. GABAergic implications in anxiety and related disorders. Biochem Biophys Res Commun 2024; 724:150218. [PMID: 38865810 DOI: 10.1016/j.bbrc.2024.150218] [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: 02/28/2024] [Revised: 05/05/2024] [Accepted: 06/02/2024] [Indexed: 06/14/2024]
Abstract
Evidence indicates that anxiety disorders arise from an imbalance in the functioning of brain circuits that govern the modulation of emotional responses to possibly threatening stimuli. The circuits under consideration in this context include the amygdala's bottom-up activity, which signifies the existence of stimuli that may be seen as dangerous. Moreover, these circuits encompass top-down regulatory processes that originate in the prefrontal cortex, facilitating the communication of the emotional significance associated with the inputs. Diverse databases (e.g., Pubmed, ScienceDirect, Web of Science, Google Scholar) were searched for literature using a combination of different terms e.g., "anxiety", "stress", "neuroanatomy", and "neural circuits", etc. A decrease in GABAergic activity is present in both anxiety disorders and severe depression. Research on cerebral functional imaging in depressive individuals has shown reduced levels of GABA within the cortical regions. Additionally, animal studies demonstrated that a reduction in the expression of GABAA/B receptors results in a behavioral pattern resembling anxiety. The amygdala consists of inhibitory networks composed of GABAergic interneurons, responsible for modulating anxiety responses in both normal and pathological conditions. The GABAA receptor has allosteric sites (e.g., α/γ, γ/β, and α/β) which enable regulation of neuronal inhibition in the amygdala. These sites serve as molecular targets for anxiolytic medications such as benzodiazepine and barbiturates. Alterations in the levels of naturally occurring regulators of these allosteric sites, along with alterations to the composition of the GABAA receptor subunits, could potentially act as mechanisms via which the extent of neuronal inhibition is diminished in pathological anxiety disorders.
Collapse
Affiliation(s)
- Indu Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pankaj Mal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Poonam Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Anushka Paul
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manish Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
31
|
Solich J, Kolasa M, Faron-Górecka A, Pabian P, Latocha K, Korlatowicz A, Dziedzicka-Wasylewska M. Modulating Stress Susceptibility and Resilience: Insights from miRNA Manipulation and Neural Mechanisms in Mice. Mol Neurobiol 2024; 61:6135-6147. [PMID: 38280111 DOI: 10.1007/s12035-024-03922-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 01/02/2024] [Indexed: 01/29/2024]
Abstract
This study explored the impact of microRNAs, specifically mmu-miR-1a-3p and mmu-miR-155-5p, on stress susceptibility and resilience in mice of different strains. Previous research had established that C57BL/6J mice were stress-susceptible, while NET-KO and SWR/J mice displayed stress resilience. These strains also exhibited variations in the serum levels of mmu-miR-1a-3p and mmu-miR-155-5p. To investigate this further, we administered antagonistic sequences (Antagomirs) targeting these microRNAs to C57/BL/6J mice and their analogs (Agomirs) to NET-KO and SWR/J mice via intracerebroventricular (i.c.v) injection. The impact of this treatment was assessed using the forced swim test. The results showed that the stress-susceptible C57/BL/6J mice could be transformed into a stress-resilient phenotype through infusion of Antagomirs. Conversely, stress-resilient mice displayed altered behavior when treated with Ago-mmu-miR-1a-3p. The study also examined the expression of mmu-miR-1a-3p in various brain regions, revealing that changes in its expression in the cerebellum (CER) were associated with the stress response. In vitro experiments with the Neuro2a cell line indicated that the Antago/Ago-miR-1a-3p and Antago/Ago-miR-155-5p treatments affected mRNAs encoding genes related to cAMP and Ca2+ signaling, diacylglycerol kinases, and phosphodiesterases. The expression changes of genes such as Dgkq, Bdnf, Ntrk2, and Pde4b in the mouse cerebellum suggested a link between cerebellar function, synaptic plasticity, and the differential stress responses observed in susceptible and resilient mice. In summary, this research highlights the role of mmu-miR-1a-3p and mmu-miR-155-5p in regulating stress susceptibility and resilience in mice and suggests a connection between these microRNAs, cerebellar function, and synaptic plasticity in the context of stress response.
Collapse
Affiliation(s)
- J Solich
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland.
| | - M Kolasa
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - A Faron-Górecka
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - P Pabian
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - K Latocha
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - A Korlatowicz
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - M Dziedzicka-Wasylewska
- Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| |
Collapse
|
32
|
Mielcarek J, Thompson JA, Appavu B, Adelson PD, Reuter-Rice K. Nursing Interventions and Intracranial Pressure Change in Pediatric Patients With Severe Traumatic Brain Injury. Dimens Crit Care Nurs 2024; 43:231-238. [PMID: 39074225 DOI: 10.1097/dcc.0000000000000656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Nursing interventions in the care of pediatric patients with severe traumatic brain injury (TBI) can have a direct effect on intracranial pressure (ICP), yet they have been largely underexplored. Early evidence is therefore needed to describe these relationships and to determine intervention that promotes neuroprotection and recovery. OBJECTIVES The aim of this study was to examine nursing interventions within the first 72 hours of pediatric severe TBI and their effects on ICP. METHOD This is a retrospective review of pediatric patients admitted for severe TBI using a quasi-experimental approach to assess nursing interventions and their association with the patients' ICP values prior to and after each intervention. RESULTS Of the 56 patients who met the inclusion criteria, 3392 intervention events (range, 31-138 events per patient) were reported. Paired t tests conducted for each intervention type found a statistically significant relationship with suctioning and percent change in ICP values (P = .045). All other interventions showed no significant differences. DISCUSSION Standard nursing interventions, specifically suctioning, in pediatric severe TBI may affect ICP and therefore neuroprotection. Further work is needed to better understand the role and timing of nursing interventions and their influence on cerebral hemodynamics so that future TBI guidelines consider nursing care and their impact on brain injury recovery.
Collapse
|
33
|
Hansson C, Hadžibajramović E, Svensson PA, Jonsdottir IH. Increased plasma levels of neuro-related proteins in patients with stress-related exhaustion: A longitudinal study. Psychoneuroendocrinology 2024; 167:107091. [PMID: 38964018 DOI: 10.1016/j.psyneuen.2024.107091] [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: 12/22/2023] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024]
Abstract
Exhaustion disorder (ED) is a stress-related disorder characterized by physical and mental symptoms of exhaustion. Recent data suggest that pathophysiological processes in the central nervous system are involved in the biological mechanisms underlying ED. The aims of this study were to investigate if plasma levels of neuro-related proteins differ between patients with ED and healthy controls, and, if so, to investigate if these differences persist over time. Using the Olink Neuro Exploratory panel, we quantified the plasma levels of 92 neuro-related proteins in 163 ED patients at the time of diagnosis (baseline), 149 patients at long-term follow-up (7-12 years later, median follow-up time 9 years and 5 months), and 100 healthy controls. We found that the plasma levels of 40 proteins were significantly higher in the ED group at baseline compared with the control group. Out of these, the plasma levels of 36 proteins were significantly lower in the ED group at follow-up compared with the same group at baseline and the plasma levels of four proteins did not significantly differ between the groups. At follow-up, the plasma levels of two proteins were significantly lower in the ED group compared with the control group. These data support the hypothesis that pathophysiological processes in the central nervous system are involved in the biological mechanisms underlying ED.
Collapse
Affiliation(s)
- Caroline Hansson
- The Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Emina Hadžibajramović
- The Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per-Arne Svensson
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingibjörg H Jonsdottir
- The Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
34
|
Wu RS, Hamden JE, Salehzadeh M, Li MX, Poudel A, Schmidt KL, Kobor MS, Soma KK. Steroid profiling in human primary teeth via liquid chromatography-tandem mass spectrometry for long-term retrospective steroid measurement. PLoS One 2024; 19:e0309478. [PMID: 39197060 PMCID: PMC11357110 DOI: 10.1371/journal.pone.0309478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 08/13/2024] [Indexed: 08/30/2024] Open
Abstract
Steroid hormones are important modulators of many physiological processes, and measurements of steroids in blood, saliva, and urine matrices are widely used to assess endocrine pathologies and stress. However, these matrices cannot be used to retrospectively assess early-life stress and developmental endocrine pathologies, because they do not integrate steroid levels over the long term. A novel biological matrix in which to measure steroids is primary teeth (or "baby teeth"). Primary teeth develop early in life and accumulate various endogenous molecules during their gradual formation. Here, we developed and validated the first assay to measure steroids in human primary teeth using liquid chromatography-tandem spectrometry (LC-MS/MS). Our assay is highly sensitive, specific, accurate, and precise. It allows for the simultaneous quantification of 17 steroids in primary teeth (16 of which have not been examined previously in primary teeth). Overall, steroid levels in primary teeth were relatively low, and 8 steroids were quantifiable. Levels of dehydroepiandrosterone, cortisol, and progesterone were the highest of the 17 steroids examined. Next, we used this assay to perform steroid profiling in primary teeth from males and females. The same 8 steroids were quantifiable, and no sex differences were found. Levels of androgens (androstenedione and testosterone) were positively correlated, and levels of glucocorticoids (cortisol, cortisone, corticosterone, 11-dehydrocorticosterone) were also positively correlated. These data demonstrate that multiple steroids can be quantified by LC-MS/MS in human primary teeth, and this method potentially provides a powerful new way to retrospectively assess early-life stress and developmental endocrine pathologies.
Collapse
Affiliation(s)
- Ruolan S. Wu
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Jordan E. Hamden
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Melody Salehzadeh
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Michael X. Li
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Asmita Poudel
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Kim L. Schmidt
- Edwin S.H. Leong Centre for Healthy Aging, University of British Columbia, Vancouver, BC, Canada
| | - Michael S. Kobor
- Edwin S.H. Leong Centre for Healthy Aging, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Kiran K. Soma
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
35
|
Valk SL, Engert V, Puhlmann L, Linz R, Caldairou B, Bernasconi A, Bernasconi N, Bernhardt BC, Singer T. Differential increase of hippocampal subfield volume after socio-affective mental training relates to reductions in diurnal cortisol. eLife 2024; 12:RP87634. [PMID: 39196261 DOI: 10.7554/elife.87634] [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] [Indexed: 08/29/2024] Open
Abstract
The hippocampus is a central modulator of the HPA-axis, impacting the regulation of stress on brain structure, function, and behavior. The current study assessed whether three different types of 3 months mental Training Modules geared towards nurturing (a) attention-based mindfulness, (b) socio-affective, or (c) socio-cognitive skills may impact hippocampal organization by reducing stress. We evaluated mental training-induced changes in hippocampal subfield volume and intrinsic functional connectivity, by combining longitudinal structural and resting-state fMRI connectivity analysis in 332 healthy adults. We related these changes to changes in diurnal and chronic cortisol levels. We observed increases in bilateral cornu ammonis volume (CA1-3) following the 3 months compassion-based module targeting socio-affective skills (Affect module), as compared to socio-cognitive skills (Perspective module) or a waitlist cohort with no training intervention. Structural changes were paralleled by relative increases in functional connectivity of CA1-3 when fostering socio-affective as compared to socio-cognitive skills. Furthermore, training-induced changes in CA1-3 structure and function consistently correlated with reductions in cortisol output. Notably, using a multivariate approach, we found that other subfields that did not show group-level changes also contributed to changes in cortisol levels. Overall, we provide a link between a socio-emotional behavioural intervention, changes in hippocampal subfield structure and function, and reductions in cortisol in healthy adults.
Collapse
Affiliation(s)
- Sofie Louise Valk
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- INM-7, FZ Jülich, Jülich, Germany
- Institute for System Neurosciences, Heinrich Heine University, Düsseldorf, Germany
| | - Veronika Engert
- Institute for Psychosocial Medicine, Psychotherapy and Psychooncology, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
- Research Group Social Stress and Family Health, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Lara Puhlmann
- Research Group Social Stress and Family Health, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Leibniz Institute for Resilience Research, Mainz, Germany
| | - Roman Linz
- Research Group Social Stress and Family Health, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Benoit Caldairou
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Andrea Bernasconi
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Neda Bernasconi
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Boris C Bernhardt
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Tania Singer
- Social Neuroscience Lab, Max Planck Society, Berlin, Germany
| |
Collapse
|
36
|
Değirmenci MD, Çalışkan H, Güneş E. Effects of chronic intermittent cold stress on anxiety-depression-like behaviors in adolescent rats. Behav Brain Res 2024; 472:115130. [PMID: 38936426 DOI: 10.1016/j.bbr.2024.115130] [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: 04/08/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Stress, which triggers numerous physiological and behavioral responses in the organism, is a significant risk factor that contributes to the development of psychiatric disorders such as depression and anxiety. This study aimed to investigate the inflammation, oxidative stress status, anxiety, and depression-like behaviors of adolescent rodents exposed to chronic intermittent cold stress. Adolescent male rats were subjected to a modified chronic intermittent cold stress model (21 days, 1 hour/day, 4 °C). Depression-like behaviors were evaluated using the sucrose preference and forced swimming tests, while anxiety-like behaviors were assessed using the open field, elevated plus maze, and light-dark box tests. We measured levels of cortisol, tumor necrosis factor-α, interleukin-1β, brain-derived natriuretic factor, reactive oxygen species, malondialdehyde, total oxidants and antioxidants, and other chemicals in the prefrontal cortex, thalamus, striatum, and hippocampus brain regions of rats using ELISA and colorimetric methods. Data were analyzed using Student's t-test and Pearson correlation analysis. After the cold stress treatment, both anxiety and depression-like behaviors increased remarkably in the subjects. Our study revealed significant changes in various brain regions among the stress-exposed subjects. Cold stress resulted in decreased BDNF levels in the prefrontal cortex and striatum (p < 0.05), increased cortisol levels in the prefrontal cortex (p < 0.05), increased IL-1β levels in the hippocampus and thalamus (p < 0.05), increased protein carbonyl levels in the striatum (p < 0.05), and decreased TAS in the prefrontal cortex and thalamus (p < 0.05). Adolescent rats exposed to cold exhibit both anxiety- and depression-like behaviors. This study observed an increase in inflammation in various brain regions, yet the responses to stress varied. Our findings suggest that adolescence is a period of heightened sensitivity to stress, which can lead to dramatic consequences.
Collapse
Affiliation(s)
| | - Hasan Çalışkan
- Balıkesir University Medicine Faculty, Physiology Department, Balıkesir, Turkey
| | - Emel Güneş
- Ankara University Medicine Faculty, Physiology Department, Ankara, Turkey
| |
Collapse
|
37
|
Milaneschi Y, Montanari S, Jansen R, Schranner D, Kastenmüller G, Arnold M, Janiri D, Sani G, Bhattacharyya S, Dehkordi SM, Dunlop B, Rush A, Penninx B, Kaddurah-Daouk R. Acylcarnitines metabolism in depression: association with diagnostic status, depression severity and symptom profile in the NESDA cohort. RESEARCH SQUARE 2024:rs.3.rs-4638158. [PMID: 39149483 PMCID: PMC11326352 DOI: 10.21203/rs.3.rs-4638158/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Acylcarnitines (ACs) are involved in bioenergetics processes that may play a role in the pathophysiology of depression. Previous genomic evidence identified four ACs potentially linked to depression risk. We carried forward these ACs and tested the association of their circulating levels with Major Depressive Disorder (MDD) diagnosis, overall depression severity and specific symptom profiles. The sample from the Netherlands Study of Depression and Anxiety included participants with current (n = 1035) or remitted (n = 739) MDD and healthy controls (n = 800). Plasma levels of four ACs (short-chain: acetylcarnitine C2 and propionylcarnitine C3; medium-chain: octanoylcarnitine C8 and decanoylcarnitine C10) were measured. Overall depression severity as well as atypical/energy-related (AES), anhedonic and melancholic symptom profiles were derived from the Inventory of Depressive Symptomatology. As compared to healthy controls, subjects with current or remitted MDD presented similarly lower mean C2 levels (Cohen's d = 0.2, p ≤ 1e-4). Higher overall depression severity was significantly associated with higher C3 levels (ß=0.06, SE = 0.02, p = 1.21e-3). No associations were found for C8 and C10. Focusing on symptom profiles, only higher AES scores were linked to lower C2 (ß=-0.05, SE = 0.02, p = 1.85e-2) and higher C3 (ß=0.08, SE = 0.02, p = 3.41e-5) levels. Results were confirmed in analyses pooling data with an additional internal replication sample from the same subjects measured at 6-year follow-up (totaling 4141 observations). Small alterations in levels of short-chain acylcarnitine levels were related to the presence and severity of depression, especially for symptoms reflecting altered energy homeostasis. Cellular metabolic dysfunctions may represent a key pathway in depression pathophysiology potentially accessible through AC metabolism.
Collapse
Affiliation(s)
| | | | - Rick Jansen
- Amsterdam UMC location Vrije Universiteit Amsterdam
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Tanner MK, Mellert SM, Fallon IP, Baratta MV, Greenwood BN. Multiple Sex- and Circuit-Specific Mechanisms Underlie Exercise-Induced Stress Resistance. Curr Top Behav Neurosci 2024. [PMID: 39080242 DOI: 10.1007/7854_2024_490] [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] [Indexed: 08/13/2024]
Abstract
Prior physical activity reduces the risk of future stress-related mental health disorders including depression, anxiety, and post-traumatic stress disorder. Rodents allowed to engage in voluntary wheel running are similarly protected from behavioral consequences of stress. The present review summarizes current knowledge on mechanisms underlying exercise-induced stress resistance. A conceptual framework involving the development (during exercise) and expression (during stress) of stress resistance from exercise is proposed. During the development of stress resistance, adaptations involving multiple exercise signals and molecular mediators occur within neural circuits orchestrating various components of the stress response, which then respond differently to stress during the expression of stress resistance. Recent data indicate that the development and expression of stress resistance from exercise involve multiple independent mechanisms that depend on sex, stressor severity, and behavioral outcome. Recent insight into the role of the prefrontal cortex in exercise-induced stress resistance illustrates these multiple mechanisms. This knowledge has important implications for the design of future experiments aimed at identifying the mechanisms underlying exercise-induced stress resistance.
Collapse
Affiliation(s)
- Margaret K Tanner
- Department of Psychology, University of Colorado Denver, Denver, CO, USA
| | - Simone M Mellert
- Department of Integrative Biology, University of Colorado Denver, Denver, CO, USA
| | - Isabella P Fallon
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | | |
Collapse
|
39
|
Jimoh-Abdulghaffaar HO, Joel IY, Jimoh OS, Ganiyu KO, Alatiba TM, Ogunyomi VO, Adebayo MS, Awoliyi VT, Agaka AO, Oyedeji AB, Kolade IA, Ojulari LS. Sex Influences Genetic Susceptibility to Depression-Like Behaviors in Chronic Unpredictable Mild Stress-Exposed Wistar Rats. Mol Neurobiol 2024:10.1007/s12035-024-04348-5. [PMID: 39012445 DOI: 10.1007/s12035-024-04348-5] [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: 02/20/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024]
Abstract
Depression is one of the most common mood disorders among psychiatric diseases. It affects about 10% of the adult population. However, its etiopathogenesis remains poorly understood. Exploring the dynamics of stress-susceptibility and resilience will help in understanding the molecular and biological mechanisms underlying the etiopathogenesis of depression. This study aimed to determine the differences and/or similarities in factors responsible for susceptibility to depression-like behaviors in male and female Wistar rats subjected to chronic unpredictable mild stress (CUMS). Sixty Wistar rats (30 male and 30 female) weighing between 120 and 150 g were used for this study. The rats were divided into two sub-groups: control (10) and test (20) groups. Rats in the test groups were subjected to CUMS. Depression-like behaviors were assessed using light-dark box, sucrose preference, and tail suspension tests. Rats that showed depression-like behaviors following the behavioral tests (CUMS-susceptible group) were sacrificed, and their hippocampi were excised. Genomic deoxyribonucleic acid (gDNA) was purified from the hippocampal samples. Purified gDNA was subjected to whole genome sequencing (WGS). Base-calling of sequence reads from raw sequencing signal (FAST5) files was carried out, and variants were called from alignment BAM files. The corresponding VCF files generated from the variant calling experiment were filtered. Genes were identified, their impacts estimated, and variants annotated. Functional enrichment analysis was then carried out. Approximately 41% of the male and 49% of the female rats subjected to CUMS showed significant (p < 0.05) depression-like behaviors following assessment on behavioral tests. WGS of the hippocampal DNA revealed 289,839 single nucleotide polymorphisms variant types, 7002 insertions, and 34,459 deletions in males, and 1,570,186 single nucleotide polymorphisms variant types, 109,860 insertions, and 597,241 deletions in female Wistar rats. Three genes with high-impact variants were identified in male and 22 in female Wistar rats, respectively. In conclusion, female Wistar rats are more susceptible to depression-like behaviors after exposure to CUMS than males. They also have more gene variants (especially high-impact variants) than male Wistar rats.
Collapse
Affiliation(s)
| | - Ireoluwa Yinka Joel
- Department of Biochemistry, Federal University of Agriculture, Makurdi, Benue State, Nigeria
| | | | - Kaosara Oyinola Ganiyu
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Temidayo Micheal Alatiba
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Victory Oluwaseyi Ogunyomi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Muhammed Salaudeen Adebayo
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Victoria Tolulope Awoliyi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Adamah Olamide Agaka
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Aminat Bolatito Oyedeji
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Ifeoluwa A Kolade
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lekan Sheriff Ojulari
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
40
|
Herzberg MP, Smyser CD. Prenatal Social Determinants of Health: Narrative review of maternal environments and neonatal brain development. Pediatr Res 2024:10.1038/s41390-024-03345-7. [PMID: 38961164 DOI: 10.1038/s41390-024-03345-7] [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] [Received: 02/01/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
The Social Determinants of Health, a set of social factors including socioeconomic status, community context, and neighborhood safety among others, are well-known predictors of mental and physical health across the lifespan. Recent research has begun to establish the importance of these social factors at the earliest points of brain development, including during the prenatal period. Prenatal socioeconomic status, perceived stress, and neighborhood safety have all been reported to impact neonatal brain structure and function, with exploratory work suggesting subsequent effects on infant and child behavior. Secondary effects of the Social Determinants of Health, such as maternal sleep and psychopathology during pregnancy, have also been established as important predictors of infant brain development. This research not only establishes prenatal Social Determinants of Health as important predictors of future outcomes but may be effectively applied even before birth. Future research replicating and extending the effects in this nascent literature has great potential to produce more specific and mechanistic understanding of the social factors that shape early neurobehavioral development. IMPACT: This review synthesizes the research to date examining the effects of the Social Determinants of Health during the prenatal period and neonatal brain outcomes. Structural, functional, and diffusion-based imaging methodologies are included along with the limited literature assessing subsequent infant behavior. The degree to which results converge between studies is discussed, in combination with the methodological and sampling considerations that may contribute to divergence in study results. Several future directions are identified, including new theoretical approaches to assessing the impact of the Social Determinants of Health during the perinatal period.
Collapse
Affiliation(s)
- Max P Herzberg
- Department of Psychiatry, Washington University in St. Louis, Saint Louis, MO, USA
| | - Christopher D Smyser
- Department of Neurology, Pediatrics, and Radiology, Washington University in St. Louis, Saint Louis, MO, USA.
| |
Collapse
|
41
|
Migeot J, Panesso C, Duran-Aniotz C, Ávila-Rincón C, Ochoa C, Huepe D, Santamaría-García H, Miranda JJ, Escobar MJ, Pina-Escudero S, Romero-Ortuno R, Lawlor B, Ibáñez A, Lipina S. Allostasis, health, and development in Latin America. Neurosci Biobehav Rev 2024; 162:105697. [PMID: 38710422 PMCID: PMC11162912 DOI: 10.1016/j.neubiorev.2024.105697] [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: 10/24/2023] [Revised: 03/05/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
The lifespan is influenced by adverse childhood experiences that create predispositions to poor health outcomes. Here we propose an allostatic framework of childhood experiences and their impact on health across the lifespan, focusing on Latin American and Caribbean countries. This region is marked by significant social and health inequalities nested in environmental and social stressors, such as exposure to pollution, violence, and nutritional deficiencies, which critically influence current and later-life health outcomes. We review several manifestations across cognition, behavior, and the body, observed at the psychological (e.g., cognitive, socioemotional, and behavioral dysfunctions), brain (e.g., alteration of the development, structure, and function of the brain), and physiological levels (e.g., dysregulation of the body systems and damage to organs). To address the complexity of the interactions between environmental and health-related factors, we present an allostatic framework regarding the cumulative burden of environmental stressors on physiological systems (e.g., cardiovascular, metabolic, immune, and neuroendocrine) related to health across the life course. Lastly, we explore the relevance of this allostatic integrative approach in informing regional interventions and public policy recommendations. We also propose a research agenda, potentially providing detailed profiling and personalized care by assessing the social and environmental conditions. This framework could facilitate the delivery of evidence-based interventions and informed childhood-centered policy-making.
Collapse
Affiliation(s)
- Joaquín Migeot
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile; Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Carolina Panesso
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Claudia Duran-Aniotz
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile; Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Cristian Ávila-Rincón
- Pontificia Universidad Javeriana (PhD Program in Neuroscience) Bogotá, San Ignacio, Colombia
| | - Carolina Ochoa
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
| | - David Huepe
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Hernando Santamaría-García
- Pontificia Universidad Javeriana (PhD Program in Neuroscience) Bogotá, San Ignacio, Colombia; Global Brain Health Institute, University of California San Francisco, San Francisco, CA, USA; Center of Memory and Cognition Intellectus, Hospital Universitario San Ignacio Bogotá, San Ignacio, Colombia
| | - J Jaime Miranda
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia; CRONICAS Centre of Excellence in Chronic Diseases, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - María Josefina Escobar
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Stefanie Pina-Escudero
- Global Brain Health Institute, Memory and Aging Center, University of California, San Francisco, USA
| | - Roman Romero-Ortuno
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; Discipline of Medical Gerontology, School of Medicine, Mercer's Institute for Successful Ageing, St James's Hospital, Dublin, Ireland
| | - Brian Lawlor
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Agustín Ibáñez
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile; Global Brain Health Institute, University of California San Francisco, San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina.
| | - Sebastián Lipina
- Unidad de Neurobiología Aplicada (UNA, CEMIC-CONICET), Buenos Aires, Argentina.
| |
Collapse
|
42
|
O'Connor AM, Hagenauer MH, Thew Forrester LC, Maras PM, Arakawa K, Hebda-Bauer EK, Khalil H, Richardson ER, Rob FI, Sannah Y, Watson SJ, Akil H. Adolescent environmental enrichment induces social resilience and alters neural gene expression in a selectively bred rodent model with anxious phenotype. Neurobiol Stress 2024; 31:100651. [PMID: 38933284 PMCID: PMC11201356 DOI: 10.1016/j.ynstr.2024.100651] [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: 10/04/2023] [Revised: 04/10/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Stress is a major influence on mental health status; the ways that individuals respond to or copes with stressors determine whether they are negatively affected in the future. Stress responses are established by an interplay between genetics, environment, and life experiences. Psychosocial stress is particularly impactful during adolescence, a critical period for the development of mood disorders. In this study we compared two established, selectively-bred Sprague Dawley rat lines, the "internalizing" bred Low Responder (bLR) line versus the "externalizing" bred High Responder (bHR) line, to investigate how genetic temperament and adolescent environment impact future responses to social interactions and psychosocial stress, and how these determinants of stress response interact. Male bLR and bHR rats were exposed to social and environmental enrichment in adolescence prior to experiencing social defeat and were then assessed for social interaction and anxiety-like behavior. Adolescent enrichment caused rats to display more social interaction, as well as nominally less social avoidance, less submission during defeat, and resilience to the effects of social stress on corticosterone, in a manner that seemed more notable in bLRs. For bHRs, enrichment also caused greater aggression during a neutral social encounter and nominally during defeat, and decreased anxiety-like behavior. To explore the neurobiology underlying the development of social resilience in the anxious phenotype bLRs, RNA-seq was conducted on the hippocampus and nucleus accumbens, two brain regions that mediate stress regulation and social behavior. Gene sets previously associated with stress, social behavior, aggression and exploratory activity were enriched with differential expression in both regions, with a particularly large effect on gene sets that regulate social behaviors. Our findings provide further evidence that adolescent enrichment can serve as an inoculating experience against future stressors. The ability to induce social resilience in a usually anxious line of animals by manipulating their environment has translational implications, as it underscores the feasibility of intervention strategies targeted at genetically vulnerable adolescent populations.
Collapse
Affiliation(s)
| | - Megan Hastings Hagenauer
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Liam Cannon Thew Forrester
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Pamela M. Maras
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Keiko Arakawa
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Elaine K. Hebda-Bauer
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Huzefa Khalil
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Evelyn R. Richardson
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Farizah I. Rob
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Yusra Sannah
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Stanley J. Watson
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Huda Akil
- Michigan Neuroscience Institute, 205 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, USA, 48109
| |
Collapse
|
43
|
Kim H, Jeong H, Lee J, Yei J, Suh M. The Effects of Acute Stress on Evoked-cortical Connectivity through Wide-field Optical Mapping of Neural and Hemodynamic Signals. Exp Neurobiol 2024; 33:140-151. [PMID: 38993081 PMCID: PMC11247281 DOI: 10.5607/en23009] [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: 03/09/2023] [Revised: 06/12/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
A single exposure to stress can induce functional changes in neurons, potentially leading to acute stress disorder or post-traumatic stress disorder. In this study, we used in vivo wide-field optical mapping to simultaneously measure neural calcium signals and hemodynamic responses over the whole cortical area. We found that cortical mapping to whisker stimuli was altered under acute stress conditions. In particular, callosal projections in the anterior cortex (primary/secondary motor, somatosensory forelimb cortex) relative to barrel field (S1BF) of somatosensory cortex were weakened. On the contrary, the projections in posterior cortex relative to S1BF were mostly unchanged or were only occasionally strengthened. In addition, changes in intra-cortical connection were opposite to those in inter-cortical connection. Thus, the S1BF connections to the anterior cortex were strengthened while those to the posterior cortex were weakened. This suggests that the well-known barrel cortex projection route was enhanced. In summary, our in vivo wide-field optical mapping study indicates that a single acute stress can impact whole-brain networks, affecting both neural and hemodynamic responses.
Collapse
Affiliation(s)
- Hayeon Kim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Haebin Jeong
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
- School of Medicine, CHA University, Seongnam 13488, Korea
| | - Jiyoung Lee
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Jaeseung Yei
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
- Department of Intelligent Precision Healthcare Convergence (IPHC), Sungkyunkwan University, Suwon 16419, Korea
| | - Minah Suh
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
- Department of Intelligent Precision Healthcare Convergence (IPHC), Sungkyunkwan University, Suwon 16419, Korea
- Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Suwon 16419, Korea
- KIST-SKKU Brain Research Center, Sungkyunkwan University, Suwon 16419, Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- IMNEWRUN Inc., Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
44
|
Nakos Bimpos M, Karali K, Antoniou C, Palermos D, Fouka M, Delis A, Tzieras I, Chrousos GP, Koutmani Y, Stefanis L, Polissidis A. Alpha-synuclein-induced stress sensitivity renders the Parkinson's disease brain susceptible to neurodegeneration. Acta Neuropathol Commun 2024; 12:100. [PMID: 38886854 PMCID: PMC11181569 DOI: 10.1186/s40478-024-01797-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/12/2024] [Indexed: 06/20/2024] Open
Abstract
A link between chronic stress and Parkinson's disease (PD) pathogenesis is emerging. Ample evidence demonstrates that the presynaptic neuronal protein alpha-synuclein (asyn) is closely tied to PD pathogenesis. However, it is not known whether stress system dysfunction is present in PD, if asyn is involved, and if, together, they contribute to neurodegeneration. To address these questions, we assess stress axis function in transgenic rats overexpressing full-length wildtype human asyn (asyn BAC rats) and perform multi-level stress and PD phenotyping following chronic corticosterone administration. Stress signaling, namely corticotropin-releasing factor, glucocorticoid and mineralocorticoid receptor gene expression, is also examined in post-mortem PD patient brains. Overexpression of human wildtype asyn leads to HPA axis dysregulation in rats, while chronic corticosterone administration significantly aggravates nigrostriatal degeneration, serine129 phosphorylated asyn (pS129) expression and neuroinflammation, leading to phenoconversion from a prodromal to an overt motor PD phenotype. Interestingly, chronic corticosterone in asyn BAC rats induces a robust, twofold increase in pS129 expression in the hypothalamus, the master regulator of the stress response, while the hippocampus, both a regulator and a target of the stress response, also demonstrates elevated pS129 asyn levels and altered markers of stress signalling. Finally, defective hippocampal stress signalling is mirrored in human PD brains and correlates with asyn expression levels. Taken together, our results link brain stress system dysregulation with asyn and provide evidence that elevated circulating glucocorticoids can contribute to asyn-induced neurodegeneration, ultimately triggering phenoconversion from prodromal to overt PD.
Collapse
Affiliation(s)
- Modestos Nakos Bimpos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Katerina Karali
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- German Center for Neurodegenerative Diseases, Feodor-Lynen-Straße 17, 81377, Munich, Germany
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, 15784, Illisia, Athens, Greece
| | - Christine Antoniou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, 15784, Illisia, Athens, Greece
| | - Dionysios Palermos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Maria Fouka
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Anastasios Delis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Iason Tzieras
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - George Panagiotis Chrousos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- University Research Institute on Maternal and Child Health and Precision Medicine, and UNESCO Chair on Adolescent Health Care, Aghia Sophia Children's Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Yassemi Koutmani
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Leonidas Stefanis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- 1St Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Alexia Polissidis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece.
- Department of Science and Mathematics, ACG-Research Center, Deree - American College of Greece, 15342, Athens, Greece.
| |
Collapse
|
45
|
Jiang Y, Gao Y, Dong D, Sun X, Situ W, Yao S. The amygdala volume moderates the relationship between childhood maltreatment and callous-unemotional traits in adolescents with conduct disorder. Eur Child Adolesc Psychiatry 2024:10.1007/s00787-024-02482-y. [PMID: 38832960 DOI: 10.1007/s00787-024-02482-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
CU traits, characterized by shallow affect, lack of fear, and absence of remorse, have been moderately associated with childhood maltreatment in a recent meta-analysis. However, the potential impact of brain structures remains undetermined. This paper examines the relationship between callous-unemotional (CU) traits, childhood maltreatment, and amygdala volumes. In this study, we used a region-of-interest (ROI) analysis to explore the interaction between the volumes of the amygdala, childhood maltreatment, and the manifestation of CU traits in adolescents diagnosed with conduct disorder (CD, N = 67), along with a comparison group of healthy-control youths (HCs, N = 89). The ROI analysis revealed no significant group differences in the bilateral amygdalar volumes. Significant positive correlation was discovered between all forms of child maltreatment (except for physical neglect) and CU traits across subjects. But the interaction of physical abuse and amygdala volumes was only significant within CD patients. Notably, a sensitivity analysis suggested that gender significantly influences these findings. These results contribute critical insights into the etiology of CU traits, emphasizing the need for customized clinical assessment tools and intervention strategies.
Collapse
Affiliation(s)
- Yali Jiang
- Department of Psychology, School of Education Science, Hunan Normal University, Changsha, People's Republic of China.
- Cognition and Human Behavior Key Laboratory of Hunan Province, Hunan Normal University, Changsha, People's Republic of China.
- Institute for Interdisciplinary Studies, Hunan Normal University, Changsha, People's Republic of China.
- Research Base for Mental Health Education of Hunan Province, Hunan Normal University, Changsha, People's Republic of China.
- School of Psychology, South China Normal University, Guangzhou, People's Republic of China.
| | - Yidian Gao
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham, UK
| | - Daifeng Dong
- Medical Psychological Center, the Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Xiaoqiang Sun
- Medical Psychological Center, the Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Weijun Situ
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Shuqiao Yao
- Medical Psychological Center, the Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
- National Clinical Research Center on Psychiatry and Psychology, Changsha, People's Republic of China
| |
Collapse
|
46
|
Nostadt A, Schlaffke L, Merz CJ, Wolf OT, Nitsche MA, Tegenthoff M, Lissek S. Microstructural differences in the cingulum and the inferior longitudinal fasciculus are associated with (extinction) learning. BMC Psychol 2024; 12:324. [PMID: 38831468 PMCID: PMC11149371 DOI: 10.1186/s40359-024-01800-y] [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: 09/15/2023] [Accepted: 05/19/2024] [Indexed: 06/05/2024] Open
Abstract
Cognitive functions, such as learning and memory processes, depend on effective communication between brain regions which is facilitated by white matter tracts (WMT). We investigated the microstructural properties and the contribution of WMT to extinction learning and memory in a predictive learning task. Forty-two healthy participants completed an extinction learning paradigm without a fear component. We examined differences in microstructural properties using diffusion tensor imaging to identify underlying neural connectivity and structural correlates of extinction learning and their potential implications for the renewal effect. Participants with good acquisition performance exhibited higher fractional anisotropy (FA) in WMT including the bilateral inferior longitudinal fasciculus (ILF) and the right temporal part of the cingulum (CNG). This indicates enhanced connectivity and communication between brain regions relevant to learning and memory resulting in better learning performance. Our results suggest that successful acquisition and extinction performance were linked to enhanced structural connectivity. Lower radial diffusivity (RD) in the right ILF and right temporal part of the CNG was observed for participants with good acquisition learning performance. This observation suggests that learning difficulties associated with increased RD may potentially be due to less myelinated axons in relevant WMT. Also, participants with good acquisition performance were more likely to show a renewal effect. The results point towards a potential role of structural integrity in extinction-relevant WMT for acquisition and extinction.
Collapse
Affiliation(s)
- Alina Nostadt
- Department of Neurology, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, 44789, Germany.
- Ruhr University Bochum, Bochum, Germany.
| | - Lara Schlaffke
- Department of Neurology, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, 44789, Germany
| | - Christian J Merz
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, 44801, Germany
| | - Oliver T Wolf
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, 44801, Germany
| | - Michael A Nitsche
- Leibniz Research Centre for Working Environment and Human Factors, Department of Psychology and Neurosciences, Dortmund, 44139, Germany
- German Centre for Mental Health (DZPG), Bochum, Germany
- University Hospital OWL, Protestant Hospital of Bethel Foundation, University Clinic of Psychiatry and Psychotherapy and University Clinic of Child and Adolescent Psychiatry and Psychotherapy, Bielefeld University, Bielefeld, 33617, Germany
| | - Martin Tegenthoff
- Department of Neurology, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, 44789, Germany
| | - Silke Lissek
- Department of Neurology, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, 44789, Germany
| |
Collapse
|
47
|
Arya H, Tamta K, Kumar A, Arya S, Maurya RC. Unpredictable chronic mild stress shows neuronal remodeling in multipolar projection neurons of hippocampal complex in postnatal chicks. Anat Sci Int 2024; 99:254-267. [PMID: 38448780 DOI: 10.1007/s12565-024-00758-6] [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: 07/26/2023] [Accepted: 02/01/2024] [Indexed: 03/08/2024]
Abstract
The hippocampal complex of birds is a narrow-curved strip of tissue that plays a crucial role in learning, memory, spatial navigation, and emotional and sexual behavior. This study was conducted to evaluate the effect of unpredictable chronic mild stress in multipolar neurons of 3-, 5-, 7-, and 9-week-old chick's hippocampal complex. This study revealed that chronic stress results in neuronal remodeling by causing alterations in dendritic field, axonal length, secondary branching, corrected spine number, and dendritic branching at 25, 50, 75, and 100 µm. Due to stress, the overall dendritic length was significantly retracted in 3-week-old chick, whereas no significant difference was observed in 5- and 7-week-old chick, but again it was significantly retracted in 9-week-old chick along with the axonal length. So, this study indicates that during initial days of stress exposure, the dendritic field shows retraction, but when the stress continues up to a certain level, the neurons undergo structural modifications so that chicks adapt and survive in stressful conditions. The repeated exposure to chronic stress for longer duration leads to the neuronal structural disruption by retraction in the dendritic length as well as axonal length. Another characteristic which leads to structural alterations is the dendritic spines which significantly decreased in all age groups of stressed chicks and eventually leads to less synaptic connections, disturbance in physiology, and neurology, which affects the learning, memory, and coping ability of an individual.
Collapse
Affiliation(s)
- Hemlata Arya
- Department of Zoology (DST-FIST Sponsored), Soban Singh Jeena University, Almora, Uttarakhand, India
- Kumaun University, Nainital, Uttarakhand, India
| | - Kavita Tamta
- Department of Zoology (DST-FIST Sponsored), Soban Singh Jeena University, Almora, Uttarakhand, India
- Kumaun University, Nainital, Uttarakhand, India
| | - Adarsh Kumar
- Department of Applied Science, Dr. K.N. Modi University, Newai-Tonk, Rajasthan, 304021, India
| | - Shweta Arya
- Department of Zoology (DST-FIST Sponsored), Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Ram Chandra Maurya
- Department of Zoology (DST-FIST Sponsored), Soban Singh Jeena University, Almora, Uttarakhand, India.
- Kumaun University, Nainital, Uttarakhand, India.
| |
Collapse
|
48
|
Domes G, Linnig K, von Dawans B. Gonads under stress: A systematic review and meta-analysis on the effects of acute psychosocial stress on gonadal steroids secretion in humans. Psychoneuroendocrinology 2024; 164:107004. [PMID: 38471257 DOI: 10.1016/j.psyneuen.2024.107004] [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: 11/02/2023] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024]
Abstract
Animal research has shown that the hypothalamus-pituitary-gonadal (HPG) axis is inhibited by (chronic and/or severe) stress, which can lead to impaired fertility and reproductive functioning, presumably caused by the inhibition of gonadal steroid secretion and in interactions with glucocorticoids. However, what has not been clarified is how acute psychosocial stress modulates gonadal steroid secretion in humans. Here we summarize the experimental research on the acute effects of stress on the secretion of gonadal steroids in humans. A systematic literature search revealed 21 studies (with N=881 individuals) measuring testosterone, progesterone or estradiol in response to a standardized acute laboratory stressor in healthy humans. Both our literature review and quantitative meta-analysis suggest that in humans, acute stress stimulates rather than inhibits HPG axis activity, although there is a considerable heterogeneity in the reported methods and results. Increased gonadal steroids in response to acute stress contrasts with many animal studies reporting the opposite pattern, at least regarding severe and/or chronic stressors. We discuss methodological issues and challenges for future research and hope to stimulate experimental studies within this area. A better understanding of these mechanisms is needed, and may have important implications for health and disease, as well as the modulation of various behaviors by acute stressors.
Collapse
Affiliation(s)
- Gregor Domes
- Department of Biological and Clinical Psychology, University of Trier, Germany; Institute for Cognitive and Affective Neuroscience, University of Trier, Germany.
| | - Katrin Linnig
- Department of Biological and Clinical Psychology, University of Trier, Germany; Institute for Cognitive and Affective Neuroscience, University of Trier, Germany
| | - Bernadette von Dawans
- Department of Biological and Clinical Psychology, University of Trier, Germany; Institute for Cognitive and Affective Neuroscience, University of Trier, Germany
| |
Collapse
|
49
|
Ruvalcaba-Delgadillo Y, Martínez-Fernández DE, Luquin S, Moreno-Alcázar A, Redolar-Ripoll D, Jauregui-Huerta F, Fernández-Quezada D. Visual EMDR stimulation mitigates acute varied stress effects on morphology of hippocampal neurons in male Wistar rats. Front Psychiatry 2024; 15:1396550. [PMID: 38803673 PMCID: PMC11129278 DOI: 10.3389/fpsyt.2024.1396550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Stress is a pervasive health concern known to induce physiological changes, particularly impacting the vulnerable hippocampus and the morphological integrity of its main residing cells, the hippocampal neurons. Eye Movement Desensitization and Reprocessing (EMDR), initially developed to alleviate emotional distress, has emerged as a potential therapeutic/preventive intervention for other stress-related disorders. This study aimed to investigate the impact of Acute Variable Stress (AVS) on hippocampal neurons and the potential protective effects of EMDR. Methods Rats were exposed to diverse stressors for 7 days, followed by dendritic morphology assessment of hippocampal neurons using Golgi-Cox staining. Results AVS resulted in significant dendritic atrophy, evidenced by reduced dendritic branches and length. In contrast, rats receiving EMDR treatment alongside stress exposure exhibited preserved dendritic morphology comparable to controls, suggesting EMDR's protective role against stressinduced dendritic remodeling. Conclusions These findings highlight the potential of EMDR as a neuroprotective intervention in mitigating stress-related hippocampal alterations.
Collapse
Affiliation(s)
- Yaveth Ruvalcaba-Delgadillo
- Neuroscience Department, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | | | - Sonia Luquin
- Neuroscience Department, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Ana Moreno-Alcázar
- ISOMAE Institute of Neurosciences and Psychosomatic Psychology, Sant Cugat del Vallés, Spain. Centre Fòrum Research Unit, Hospital del Mar, Barcelona, Spain
| | | | - Fernando Jauregui-Huerta
- Laboratorio de Fisiología del Comportamiento, Departamento de Fisiología, Facultad de medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - David Fernández-Quezada
- Neuroscience Department, University Center of Health Sciences, University of Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
50
|
Scarpa G, Antonoudiou P, Weiss G, Stone B, Maguire JL. Sex-dependent effects of early life stress on network and behavioral states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593547. [PMID: 38766016 PMCID: PMC11100797 DOI: 10.1101/2024.05.10.593547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Background Adverse childhood experiences (ACEs) are associated with numerous detriments in health, including increased vulnerability to psychiatric illnesses. Early life stress (ELS) in rodents has been shown to effectively model several of the behavioral and endocrine impacts of ACEs and has been utilized to investigate the underlying mechanisms contributing to disease. However, the precise neural mechanisms responsible for mediating the impact of ELS on vulnerability to psychiatric illnesses remain largely unknown. Methods We use behavior, immunoassay, in vivo LFP recording, histology, and patch clamp to describe the effects of ELS on stress behaviors, endocrinology, network states, protein expression, and cellular physiology in male and female mice. Results We demonstrate that a murine maternal separation (MS) ELS model causes sex-dependent alterations in behavioral and hormonal responses following an acute stressor. Local field potential (LFP) recordings in the basolateral amygdala (BLA) and frontal cortex (FC) reveal similar sex-dependent alterations at baseline, in response to acute ethological stress, and during fear memory extinction, supporting a large body of literature demonstrating that these network states contribute to stress reactivity and vulnerability to psychiatric illnesses. Sex differences were accompanied by altered physiology of BLA principal neurons in males and BLA PV interneurons in females. Conclusions Collectively, these results implicate novel, sex-dependent mechanisms through which ACEs may impact psychiatric health, involving altered cellular physiology and network states involved in emotional processing.
Collapse
|