1
|
Nalepa IF, Nielsen V, Wolf TE, Touma C, Grupe M, Asuni AA, Ratner C. Sex differences in the murine HPA axis after acute and repeated restraint stress. Stress 2025; 28:2447079. [PMID: 39819340 DOI: 10.1080/10253890.2024.2447079] [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: 07/04/2024] [Accepted: 12/08/2024] [Indexed: 01/19/2025] Open
Abstract
Chronic stress and stress-related mental illnesses such as major depressive disorder (MDD) constitute some of the leading causes of disability worldwide with a higher prevalence in women compared to men. However, preclinical research into stress and MDD is heavily biased toward using male animals only. Aberrant activity of the hypothalamic-pituitary-adrenal (HPA) axis has been linked to the development of MDD and several animal models of MDD have been established based on HPA axis dysregulation. In the present study, we compared stress biomarkers and behavior of male and female mice after acute and chronic restraint stress to investigate potential effects of sex differences in the stress response. Further, the validity of the interrupted repeated restraint stress (IRRS) model as an animal model for the HPA axis disturbances seen in MDD was assessed. After acute stress, female mice showed increased corticosterone secretion and changes in molecular markers suggesting increased HPA axis feedback sensitivity. Acute stress-induced signs of anxiety-like behavior were observed in male mice only suggesting that female mice may be more resilient to the anxiogenic effects of acute stress. Males and females responded similarly to IRRS with no sustained perturbations in HPA axis biomarkers. The IRRS model did not adequately translate to the changes reported in MDD with HPA axis overactivity and more severe perturbation models are likely needed. However, in alignment with previous studies, these data support that there are important sex differences in the HPA axis and that these may contribute to the etiology of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Isabella Flor Nalepa
- Department of Preclinical Fluid Biomarkers & Occupancy, H. Lundbeck A/S, Valby, Denmark
| | - Vibeke Nielsen
- Department of Preclinical Fluid Biomarkers & Occupancy, H. Lundbeck A/S, Valby, Denmark
| | | | - Chadi Touma
- Osnabrück University, Behavioural Biology, Osnabrück, Germany
| | - Morten Grupe
- Department of Symptom Biology, H. Lundbeck A/S, Valby, Denmark
| | - Ayodeji A Asuni
- Department of Preclinical Fluid Biomarkers & Occupancy, H. Lundbeck A/S, Valby, Denmark
| | - Cecilia Ratner
- Department of Preclinical Fluid Biomarkers & Occupancy, H. Lundbeck A/S, Valby, Denmark
| |
Collapse
|
2
|
Silva JP, Corrales WA, Catalán J, Olave FA, González-Mori PI, Alarcón M, Guarnieri T, Aliaga E, Maracaja-Coutinho V, Fiedler JL. Comprehensive Analysis of circRNA Expression and circRNA-miRNA-mRNA Networks in the Ventral Hippocampus of the Rat: Impact of Chronic Stress and Biological Sex. ACS Chem Neurosci 2025. [PMID: 40257053 DOI: 10.1021/acschemneuro.4c00681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025] Open
Abstract
This study provides new insights into how sex and chronic stress influence circRNA expression in the rat ventral hippocampus, a region critical for emotional processing. We identified 206 sex-biased circRNAs and 194 stress-responsive circRNAs, highlighting distinct expression profiles. Parental genes of male circRNAs were primarily enriched in synaptic transmission pathways, while those of female circRNAs were associated with axon guidance, emphasizing sex-specific molecular differences. Chronic stress also triggered miRNA changes unique to each sex, revealing divergent regulatory mechanisms. The identified circRNA-miRNA-mRNA axes, modulated under stress, appear to regulate the translation of numerous potential mRNA targets. In males, stress positively regulated neuroprotective pathways, suggesting a compensatory response to mitigate stress-induced damage. In contrast, females exhibited a broader translational network that favored mRNA expression without distinct pathway-specific actions. However, the smaller repressed network in females─characterized by a higher circRNA-to-miRNA and mRNA ratio─may indicate a more selective and targeted regulatory mechanism, with many interactions linked to anti-inflammatory processes. Coexpression analysis revealed two male-specific modules with altered activity under stress. These were associated with processes such as reticulum stress and actin dynamics, the latter linked to dendritic spine loss and depressive-like behaviors, extensively documented in chronically stressed male rats. Conversely, females displayed an activated stress-responsive module, promoting axon guidance and long-term potentiation, which may contribute to improved cognitive outcomes. Among the identified circRNAs, rno-Gabrg3_0001 emerged as stress-sensitive in males. This circRNA exhibited predicted miRNA binding sites and interactions with proteins involved in vesicle trafficking, forming part of a highly active module enriched in genes related to ion transport and membrane protein localization. Overall, these findings uncover sex-dependent regulatory mechanisms driving transcriptomic changes under chronic stress, deepening our understanding of ventral hippocampal molecular functions. Investigating these regulatory networks, which differentially affect the male and female ventral hippocampus, could inform the development of sex-specific therapeutic strategies for stress-related disorders.
Collapse
Affiliation(s)
- Juan Pablo Silva
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
- Unidad de Genómica Avanzada─UGA, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
| | - Wladimir A Corrales
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
- Unidad de Genómica Avanzada─UGA, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
| | - Julia Catalán
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
| | - Felipe A Olave
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
| | - Pablo I González-Mori
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
| | - Matías Alarcón
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
| | - Tatiana Guarnieri
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
| | - Esteban Aliaga
- School of Medícal Technology and The Neuropsychology and Cognitive Neurosciences Research Center (CINPSI-Neurocog), Faculty of Health Sciences, Universidad Católica del Maule, Talca 3460000, Chile
| | - Vinicius Maracaja-Coutinho
- Unidad de Genómica Avanzada─UGA, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
- Advanced Center for Chronic Diseases─ACCDiS, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
- Centro de Modelamiento Molecular, Biofísica y Bioinformática─CM2B2, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
| | - Jenny L Fiedler
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia 8380492, Santiago, Chile
| |
Collapse
|
3
|
Soares AR, Garcia-Rivas V, Fai C, Thomas M, Zheng X, Picciotto MR, Mineur YS. Sex differences in the microglial response to stress and chronic alcohol exposure in mice. Biol Sex Differ 2025; 16:19. [PMID: 40038827 DOI: 10.1186/s13293-025-00701-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Women are more susceptible to stress-induced alcohol drinking, and preclinical data suggest that stress can increase alcohol intake in female rodents; however, a comprehensive understanding of the neurobiological processes underlying this sex difference is still emerging. Neuroimmune signaling, particularly by microglia, the brain's macrophages, is known to contribute to dysregulation of limbic circuits following stress and alcohol exposure. Females exhibit heightened immune reactivity, so we set out to characterize sex differences in the microglial response to stress and alcohol exposure. METHODS Male and female C57BL/6J mice were administered alcohol over 15 or 22 trials of a modified Drinking in the Dark paradigm, with repeated exposure to inescapable footshock stress and the stress-paired context. Mice were perfused immediately after drinking and we performed immunohistochemical analyses of microglial density, morphology, and protein expression in subregions of the amygdala and hippocampus. RESULTS We observed dynamic sex differences in microglial phenotypes at baseline and in response to stress and alcohol. Microglia in the hippocampus displayed more prominent sex differences and heightened reactivity to stress and alcohol. Chronic alcohol exposure decreased density of amygdala microglia and lysosomal expression. CONCLUSION We analyzed multiple measures of microglial activation, resulting in a comprehensive assessment of microglial changes mediated by sex, stress, and alcohol. These findings highlight the complexity of microglial contributions to the development of AUD and comorbid mood and stress disorders in men and women.
Collapse
Affiliation(s)
- Alexa R Soares
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA
| | - Vernon Garcia-Rivas
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Caroline Fai
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Merrilee Thomas
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Xiaoying Zheng
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA.
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA.
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street - 3rd Floor Research, New Haven, CT, 06508, USA.
| | - Yann S Mineur
- Department of Psychiatry, Yale University, 34 Park Street, 3rd Floor Research, New Haven, CT, 06508, USA
| |
Collapse
|
4
|
Lyons CE, Pallais JP, McGonigle S, Mansk RP, Collinge CW, Yousefzadeh MJ, Baker DJ, Schrank PR, Williams JW, Niedernhofer LJ, van Deursen JM, Razzoli M, Bartolomucci A. Chronic social stress induces p16-mediated senescent cell accumulation in mice. NATURE AGING 2025; 5:48-64. [PMID: 39528642 DOI: 10.1038/s43587-024-00743-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Life stress can shorten lifespan and increase risk for aging-related diseases, but the biology underlying this phenomenon remains unclear. Here we assessed the effect of chronic stress on cellular senescence-a hallmark of aging. Exposure to restraint stress, a psychological non-social stress model, increased p21Cip1 exclusively in the brains of male, but not female mice, and in a p16Ink4a-independent manner. Conversely, exposure to chronic subordination stress (only males were tested) increased key senescent cell markers in peripheral blood mononuclear cells, adipose tissue and brain, in a p16Ink4a-dependent manner. p16Ink4a-positive cells in the brain of chronic subordination stress-exposed mice were primarily hippocampal and cortical neurons with evidence of DNA damage that could be reduced by p16Ink4a cell clearance. Clearance of p16Ink4a-positive cells was not sufficient to ameliorate the adverse effects of social stress on measured metrics of healthspan. Overall, our findings indicate that social stress induces an organ-specific and p16Ink4a-dependent accumulation of senescent cells, illuminating a fundamental way by which the social environment can contribute to aging.
Collapse
Affiliation(s)
- Carey E Lyons
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Jean Pierre Pallais
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Seth McGonigle
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Rachel P Mansk
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Charles W Collinge
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Matthew J Yousefzadeh
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Paul F. Glenn Center for the Biology of Aging, Mayo Clinic, Rochester, MN, USA
| | - Patricia R Schrank
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Jesse W Williams
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Campbell HM, Guo JD, Kuhn CM. Applying the Research Domain Criteria to Rodent Studies of Sex Differences in Chronic Stress Susceptibility. Biol Psychiatry 2024; 96:848-857. [PMID: 38821193 DOI: 10.1016/j.biopsych.2024.05.016] [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: 10/10/2023] [Revised: 04/27/2024] [Accepted: 05/17/2024] [Indexed: 06/02/2024]
Abstract
Women have a 2-fold increased rate of stress-associated psychiatric disorders such as depression and anxiety, but the mechanisms that underlie this increased susceptibility remain incompletely understood. Historically, female subjects were excluded from preclinical studies and clinical trials. Additionally, chronic stress paradigms used to study psychiatric pathology in animal models were developed for use in males. However, recent changes in National Institutes of Health policy encourage inclusion of female subjects, and considerable work has been performed in recent years to understand biological sex differences that may underlie differences in susceptibility to chronic stress-associated psychiatric conditions. Here, we review the utility as well as current challenges of using the framework of the National Institute of Mental Health's Research Domain Criteria as a transdiagnostic approach to study sex differences in rodent models of chronic stress including recent progress in the study of sex differences in the neurobehavioral domains of negative valence, positive valence, cognition, social processes, and arousal.
Collapse
Affiliation(s)
- Hannah M Campbell
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - Jessica D Guo
- Duke University School of Medicine, Durham, North Carolina
| | - Cynthia M Kuhn
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
6
|
Asle-Rousta M, Peirovy Y. Neuroprotective Effects of Thymol and p-Cymene in Immobilized Male rats through Alterations in Molecular, Biochemical, Histological, and Behavioral Parameters. Neurochem Res 2024; 50:5. [PMID: 39540984 DOI: 10.1007/s11064-024-04271-0] [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: 07/17/2024] [Revised: 09/02/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024]
Abstract
The research was conducted to examine the neuroprotective effect of thymol and its precursor p-cymene on chronic immobility stress in adult male Wistar rats. The rats were subjected to 2.5 h of stress every day for 14 consecutive days by placing them inside a restrainer. Thymol (10 mg/kg) and p-cymene (50 mg/kg) were given to the rats during the same period. The results showed that thymol and p-cymene prevented the increase of MDA level, decline of GSH level, and decrease of SOD and GPx activity in the hippocampus of rats exposed to stress. These monoterpenes also prevented the increase in the expression of Tnfa, Il1b, Tlr4, and Nfkb, and the decrease in the expression of Nrf2, Ho1, and Bdnf. In addition, thymol and p-cymene inhibited the increase in the expression and activity of acetylcholinesterase in the hippocampus of animals exposed to immobility and enhanced the expression of A7nachr. They also reduced neuronal death in the CA1 region of stressed animals and improved their performance in the Morris water maze and elevated plus maze tests. Based on these findings, thymol and p-cymene may be effective in preventing neurodegenerative diseases as they reduce oxidative stress and neuroinflammation, strengthen ACh signaling, and stimulate Bdnf expression.
Collapse
Affiliation(s)
| | - Yasaman Peirovy
- Nanobiotechnology Research Center, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| |
Collapse
|
7
|
Goh JY, Rueda P, Taylor J, Rathbone A, Scott D, Langmead CJ, Fone KC, Stewart GD, King MV. Transcriptomic analysis of rat prefrontal cortex following chronic stress induced by social isolation - Relevance to psychiatric and neurodevelopmental illness, and implications for treatment. Neurobiol Stress 2024; 33:100679. [PMID: 39502833 PMCID: PMC11536066 DOI: 10.1016/j.ynstr.2024.100679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
Social isolation is an established risk factor for psychiatric illness, and became increasingly topical with the spread of SARS-CoV-2. We used RNA sequencing (RNA-Seq) to enable unbiased assessment of transcriptomic changes within the prefrontal cortex (PFC) of isolation-reared rats. To provide insight into the relevance of this manipulation for studying human illness, we compared differentially expressed genes (DEGs) and enriched biological functions against datasets involving post-mortem frontal cortical tissue from patients with psychiatric and neurodevelopmental illnesses. Sixteen male Sprague-Dawley rats were reared in groups of four or individually from weaning on postnatal day (PND) 22-24 until PFC tissue collection for RNA-Seq (PND64-66). We identified a total of 183 DEGs in isolates, of which 128 mirrored those in PFC tissue from patients with stress-related mental illnesses and/or neurodevelopmental conditions featuring social deficits. Seventy-one encode proteins classed as druggable by the gene-drug interaction database. Interestingly there are antagonists or inhibitors for the products of three of these up-regulated DEGs (Hrh3, Snca and Sod1) and agonists or activators for products of six of these down-regulated DEGs (Chrm4, Klf2, Lrrk2, Nr4a1, Nr4a3 and Prkca). Some have already undergone pre-clinical and clinical evaluation, and studies with the remainder may be warranted. Changes to Hrh3, Sod1, Chrm4, Lrrk2, Nr4a1 and Prkca were replicated in an independent cohort of sixteen male Sprague-Dawley rats via quantitative reverse transcription polymerase chain reaction (qRT-PCR). Our findings support the continued use of post-weaning isolation rearing to investigate the neurobiology of stress-related disorders and evaluate therapeutic targets.
Collapse
Affiliation(s)
- Jen-Yin Goh
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Patricia Rueda
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Joy Taylor
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Alex Rathbone
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Daniel Scott
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Christopher J. Langmead
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Kevin C.F. Fone
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Gregory D. Stewart
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Madeleine V. King
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| |
Collapse
|
8
|
Peirovy Y, Asle-Rousta M. Thymol and p-Cymene Protect the Liver by Mitigating Oxidative Stress, Suppressing TNF-α/NF-κB, and Enhancing Nrf2/HO-1 Expression in Immobilized Rats. Chem Biol Drug Des 2024; 104:e14618. [PMID: 39313485 DOI: 10.1111/cbdd.14618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/29/2024] [Accepted: 08/17/2024] [Indexed: 09/25/2024]
Abstract
This study aimed to investigate the effects of the monoterpenes thymol and p-cymene on the liver of rats subjected to prolonged immobilization stress and to discover the possible mechanism behind this effect. For 14 consecutive days, the rats were placed in a restrainer for 2.5 h every day to expose them to stress. During the same period, thymol (10 mg/kg, gavage) and p-cymene (50 mg/kg, intraperitoneally) were also administered. Thymol and p-cymene prevented the increase in malondialdehyde levels and the decrease in glutathione content in the liver of rats exposed to chronic immobility. They also increased the activity of the glutathione peroxidase enzyme in the liver of stressed animals, but only thymol could increase the activity of superoxide dismutase. These monoterpenes reduced the expression of pro-inflammatory cytokines tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, and IL-6 and nuclear factor kappa B (NF-κB) in the liver of stressed animals. They increased the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Thymol and p-cymene greatly prevented the infiltration of inflammatory cells in the liver parenchyma of stressed rats. In conclusion, the study found that thymol and p-cymene have a hepatoprotective effect on immobilized rats, likely exerted by suppressing oxidative stress and inflammation, stimulating Nrf2/HO-1 signaling, and inhibiting the TNF-α/NF-κB pathway.
Collapse
Affiliation(s)
- Yasaman Peirovy
- Nanobiotechnology Research Center, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | | |
Collapse
|
9
|
Krolick KN, Cao J, Gulla EM, Bhardwaj M, Marshall SJ, Zhou EY, Kiss AJ, Choueiry F, Zhu J, Shi H. Subregion-specific transcriptomic profiling of rat brain reveals sex-distinct gene expression impacted by adolescent stress. Neuroscience 2024; 553:19-39. [PMID: 38977070 PMCID: PMC11444371 DOI: 10.1016/j.neuroscience.2024.07.002] [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: 01/18/2024] [Revised: 05/14/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Stress during adolescence clearly impacts brain development and function. Sex differences in adolescent stress-induced or exacerbated emotional and metabolic vulnerabilities could be due to sex-distinct gene expression in hypothalamic, limbic, and prefrontal brain regions. However, adolescent stress-induced whole-genome expression changes in key subregions of these brain regions were unclear. In this study, female and male adolescent Sprague Dawley rats received one-hour restraint stress daily from postnatal day (PD) 32 to PD44. Corticosterone levels, body weights, food intake, body composition, and circulating adiposity and sex hormones were measured. On PD44, brain and blood samples were collected. Using RNA-sequencing, sex-specific differences in stress-induced differentially expressed (DE) genes were identified in subregions of the hypothalamus, limbic system, and prefrontal cortex. Canonical pathways reflected well-known sex-distinct maladies and diseases, substantiating the therapeutic potential of the DE genes found in the current study. Thus, we proposed specific sex distinct, adolescent stress-induced transcriptional changes found in the current study as examples of the molecular bases for sex differences witnessed in stress induced or exacerbated emotional and metabolic disorders. Future behavioral studies and single-cell studies are warranted to test the implications of the DE genes identified in this study in sex-distinct stress-induced susceptibilities.
Collapse
Affiliation(s)
| | - Jingyi Cao
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Evelyn M Gulla
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Meeta Bhardwaj
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | | | - Ethan Y Zhou
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Andor J Kiss
- Center for Bioinformatics & Functional Genomics, Miami University, Oxford, OH 45056, USA.
| | - Fouad Choueiry
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA.
| | - Jiangjiang Zhu
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
10
|
Poitras M, Lebeau M, Plamondon H. The cycle of stress: A systematic review of the impact of chronic psychological stress models on the rodent estrous cycle. Neurosci Biobehav Rev 2024; 162:105730. [PMID: 38763179 DOI: 10.1016/j.neubiorev.2024.105730] [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: 03/13/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Stress is known to impair reproduction through interactions between the hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-gonadal (HPG) axes. However, while it is well accepted that stress can alter estrous cycle regularity, a key indicator of female's HPG axis function, effects of different types of psychological stress have been inconsistent. This systematic review evaluated the impact of rodent models of psychological stress on estrous cyclicity, while reporting biological parameters pertaining to HPA or HPG axis function assessed within these studies. We performed a systematic database search and included articles that implemented a psychological stress model in rodents and reported estrous cyclicity for at least two cycles after initiation of stress. Of the 32 studies included, 62.5% reported post-stress alterations to estrous cyclicity, with Chronic Mild Stress (CMS) models showing the most conclusive effects. Twenty-five studies measured HPG or HPA axis markers, with cycle disruptions being commonly observed in parallel with altered estradiol and increased corticosterone levels. Our review highlights gaps in reporting estrous cyclicity assessments and makes recommendations to improve comparability between studies.
Collapse
Affiliation(s)
- Marilou Poitras
- Cerebro Vascular Accidents and Behavioural Recovery Laboratory, School of Psychology, University of Ottawa, Ottawa, Canada
| | - Madison Lebeau
- Cerebro Vascular Accidents and Behavioural Recovery Laboratory, School of Psychology, University of Ottawa, Ottawa, Canada
| | - Hélène Plamondon
- Cerebro Vascular Accidents and Behavioural Recovery Laboratory, School of Psychology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
11
|
Soares AR, Garcia-Rivas V, Fai C, Thomas MA, Zheng X, Picciotto MR, Mineur YS. Role of microglia in stress-induced alcohol intake in female and male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597614. [PMID: 38895217 PMCID: PMC11185719 DOI: 10.1101/2024.06.05.597614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Rates of alcohol use disorder (AUD) have escalated in recent years, with a particular increase among women. Women are more susceptible to stress-induced alcohol drinking, and preclinical data suggest that stress can increase alcohol intake in female rodents; however, a comprehensive understanding of sex-specific neurobiological substrates underlying this phenomenon is still emerging. Microglia, the resident macrophages of the brain, are essential for reshaping neuronal processes, and microglial activity contributes to overall neuronal plasticity. We investigated microglial dynamics and morphology in limbic brain structures of male and female mice following exposure to stress, alcohol or both challenges. In a modified paradigm of intermittent binge drinking (repeated "drinking in the dark"), we determined that female, but not male, mice increased their alcohol consumption after exposure to a physical stressor and re-exposure trials in the stress-paired context. Ethanol (EtOH) drinking and stress altered a number of microglial parameters, including overall number, in subregions of the amygdala and hippocampus, with effects that were somewhat more pronounced in female mice. We used the CSF1R antagonist PLX3397 to deplete microglia in female mice to determine whether microglia contribute to stress-induced escalation of EtOH intake. We observed that microglial depletion attenuated stress-induced alcohol intake with no effect in the unstressed group. These findings suggest that microglial activity can contribute to alcohol intake under stressful conditions, and highlight the importance of evaluating sex-specific mechanisms that could result in tailored interventions for AUD in women.
Collapse
|
12
|
Scanes CG, Pierzchała-Koziec K, Gajewska A. Effects of Restraint Stress on Circulating Corticosterone and Met Enkephalin in Chickens: Induction of Shifts in Insulin Secretion and Carbohydrate Metabolism. Animals (Basel) 2024; 14:752. [PMID: 38473137 DOI: 10.3390/ani14050752] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
This study examined the effects of acute restraint stress in the presence or absence of naltrexone on the circulating concentrations of insulin, glucose, Met-enkephalin and corticosterone in 14-week-old chickens [design: 2 sex × 2 stress/non-stress × 2 +/- naltrexone]. In chickens (five male and five females per treatment) subjected to restraint for 30 min, there were increases in the plasma concentrations of corticosterone and Met-enkephalin. The plasma concentrations of insulin and glucose were also increased in the chickens during restraint. Moreover, there were increases in the plasma concentrations of insulin and glucose in the chickens. The patterns of expression of the proenkephalin gene (PENK) in both the anterior pituitary gland and the adrenal gland were very similar to that of plasma Met-enkephalin. There were relationships between the plasma concentrations of corticosterone, Met-enkephalin, insulin and glucose after 30 min of restraint. The effects of naltrexone treatment on both untreated and stressed chickens were also examined, with naltrexone attenuating the stress-induced increases in the plasma concentrations of corticosterone, Met-enkephalin and glucose but not in those of insulin. The present study demonstrates that stress increases insulin secretion in chickens but also induces insulin resistance.
Collapse
Affiliation(s)
- Colin G Scanes
- Department of Biological Science, University of Wisconsin Milwaukee, Milwaukee, WI 53211, USA
| | - Krystyna Pierzchała-Koziec
- Department of Animal Physiology and Endocrinology, University of Agriculture, Mickiewicza 24/28, 30-059 Kraków, Poland
| | - Alina Gajewska
- Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jablonna, Poland
| |
Collapse
|
13
|
Dalla C, Jaric I, Pavlidi P, Hodes GE, Kokras N, Bespalov A, Kas MJ, Steckler T, Kabbaj M, Würbel H, Marrocco J, Tollkuhn J, Shansky R, Bangasser D, Becker JB, McCarthy M, Ferland-Beckham C. Practical solutions for including sex as a biological variable (SABV) in preclinical neuropsychopharmacological research. J Neurosci Methods 2024; 401:110003. [PMID: 37918446 PMCID: PMC10842858 DOI: 10.1016/j.jneumeth.2023.110003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/13/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Recently, many funding agencies have released guidelines on the importance of considering sex as a biological variable (SABV) as an experimental factor, aiming to address sex differences and avoid possible sex biases to enhance the reproducibility and translational relevance of preclinical research. In neuroscience and pharmacology, the female sex is often omitted from experimental designs, with researchers generalizing male-driven outcomes to both sexes, risking a biased or limited understanding of disease mechanisms and thus potentially ineffective therapeutics. Herein, we describe key methodological aspects that should be considered when sex is factored into in vitro and in vivo experiments and provide practical knowledge for researchers to incorporate SABV into preclinical research. Both age and sex significantly influence biological and behavioral processes due to critical changes at different timepoints of development for males and females and due to hormonal fluctuations across the rodent lifespan. We show that including both sexes does not require larger sample sizes, and even if sex is included as an independent variable in the study design, a moderate increase in sample size is sufficient. Moreover, the importance of tracking hormone levels in both sexes and the differentiation between sex differences and sex-related strategy in behaviors are explained. Finally, the lack of robust data on how biological sex influences the pharmacokinetic (PK), pharmacodynamic (PD), or toxicological effects of various preclinically administered drugs to animals due to the exclusion of female animals is discussed, and methodological strategies to enhance the rigor and translational relevance of preclinical research are proposed.
Collapse
Affiliation(s)
- Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Ivana Jaric
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Pavlina Pavlidi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Georgia E Hodes
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24060, USA
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece; First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Anton Bespalov
- Partnership for Assessment and Accreditation of Scientific Practice (PAASP GmbH), Heidelberg, Germany
| | - Martien J Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | | | - Mohamed Kabbaj
- Department of Biomedical Sciences & Neurosciences, College of Medicine, Florida State University, USA
| | - Hanno Würbel
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Jordan Marrocco
- Department of Biology, Touro University, New York, NY 10027, USA
| | | | - Rebecca Shansky
- Department of Psychology, Northeastern University, Boston, MA 02128, USA
| | - Debra Bangasser
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Jill B Becker
- Department of Psychology and Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margaret McCarthy
- University of Maryland School of Medicine, Department of Pharmacology, Baltimore MD, USA
| | | |
Collapse
|
14
|
Szalanczy AM, Giorgio G, Goff E, Seshie O, Grzybowski M, Klotz J, Geurts AM, Redei EE, Solberg Woods LC. Changes in environmental stress over COVID-19 pandemic likely contributed to failure to replicate adiposity phenotype associated with Krtcap3. Physiol Genomics 2023; 55:452-467. [PMID: 37458463 PMCID: PMC10642928 DOI: 10.1152/physiolgenomics.00019.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/25/2023] [Accepted: 07/09/2023] [Indexed: 07/28/2023] Open
Abstract
We previously identified keratinocyte-associated protein 3, Krtcap3, as an obesity-related gene in female rats where a whole body Krtcap3 knockout (KO) led to increased adiposity compared to wild-type (WT) controls when fed a high-fat diet (HFD). We sought to replicate this work to better understand the function of Krtcap3 but were unable to reproduce the adiposity phenotype. In the current work, WT female rats ate more compared to WT in the prior study, with corresponding increases in body weight and fat mass, while there were no changes in these measures in KO females between the studies. The prior study was conducted before the COVID-19 pandemic, while the current study started after initial lockdown orders and was completed during the pandemic in a generally less stressful environment. We hypothesize that the environmental changes impacted stress levels and may explain the failure to replicate our results. Analysis of corticosterone (CORT) at euthanasia showed a significant study-by-genotype interaction where WT had significantly higher CORT relative to KO in study 1, with no differences in study 2. These data suggest that decreasing Krtcap3 expression may alter the environmental stress response to influence adiposity. We also found that KO rats in both studies, but not WT, experienced a dramatic increase in CORT after their cage mate was removed, suggesting a separate connection to social behavioral stress. Future work is necessary to confirm and elucidate the finer mechanisms of these relationships, but these data indicate the possibility of Krtcap3 as a novel stress gene.NEW & NOTEWORTHY Obesity is linked to both genetics and environmental factors such as stress. Krtcap3 has previously been identified as a gene associated with adiposity, and our work here demonstrates that environmental stress may influence the role of Krtcap3 on both food intake and adiposity. Obesity is strongly influenced by stress in humans, so the identification of novel genes that link stress and obesity will greatly advance our understanding of the disease.
Collapse
Affiliation(s)
- Alexandria M Szalanczy
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| | - Gina Giorgio
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| | - Emily Goff
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| | - Osborne Seshie
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| | - Michael Grzybowski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jason Klotz
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Eva E Redei
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Leah C Solberg Woods
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| |
Collapse
|
15
|
Bjornson KJ, Vanderplow AM, Yang Y, Anderson DR, Kermath BA, Cahill ME. Stress-mediated dysregulation of the Rap1 small GTPase impairs hippocampal structure and function. iScience 2023; 26:107566. [PMID: 37664580 PMCID: PMC10470260 DOI: 10.1016/j.isci.2023.107566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/15/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
The effects of repeated stress on cognitive impairment are thought to be mediated, at least in part, by reductions in the stability of dendritic spines in brain regions critical for proper learning and memory, including the hippocampus. Small GTPases are particularly potent regulators of dendritic spine formation, stability, and morphology in hippocampal neurons. Through the use of small GTPase protein profiling in mice, we identify increased levels of synaptic Rap1 in the hippocampal CA3 region in response to escalating, intermittent stress. We then demonstrate that increased Rap1 in the CA3 is sufficient in and of itself to produce stress-relevant dendritic spine and cognitive phenotypes. Further, using super-resolution imaging, we investigate how the pattern of Rap1 trafficking to synapses likely underlies its effects on the stability of select dendritic spine subtypes. These findings illuminate the involvement of aberrant Rap1 regulation in the hippocampus in contributing to the psychobiological effects of stress.
Collapse
Affiliation(s)
- Kathryn J. Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amanda M. Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yezi Yang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Danielle R. Anderson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Bailey A. Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael E. Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
16
|
Szalanczy AM, Giorgio G, Goff E, Seshie O, Grzybowski M, Klotz J, Geurts AM, Redei EE, Solberg Woods LC. Changes in Environmental Stress over COVID-19 Pandemic Likely Contributed to Failure to Replicate Adiposity Phenotype Associated with Krtcap3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532439. [PMID: 36993361 PMCID: PMC10055176 DOI: 10.1101/2023.03.15.532439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
We previously identified Keratinocyte-associated protein 3, Krtcap3, as an obesity-related gene in female rats where a whole-body Krtcap3 knock-out (KO) led to increased adiposity compared to wild-type (WT) controls when fed a high-fat diet (HFD). We sought to replicate this work to better understand the function of Krtcap3 but were unable to reproduce the adiposity phenotype. In the current work, WT female rats ate more compared to WT in the prior study, with corresponding increases in body weight and fat mass, while there were no changes in these measures in KO females between the studies. The prior study was conducted before the COVID-19 pandemic, while the current study started after initial lock-down orders and was completed during the pandemic with a generally less stressful environment. We hypothesize that the environmental changes impacted stress levels and may explain the failure to replicate our results. Analysis of corticosterone (CORT) at euthanasia showed a significant study by genotype interaction where WT had significantly higher CORT relative to KO in Study 1, with no differences in Study 2. These data suggest that decreasing Krtcap3 expression may alter the environmental stress response to influence adiposity. We also found that KO rats in both studies, but not WT, experienced a dramatic increase in CORT after their cage mate was removed, suggesting a separate connection to social behavioral stress. Future work is necessary to confirm and elucidate the finer mechanisms of these relationships, but these data indicate the possibility of Krtcap3 as a novel stress gene.
Collapse
Affiliation(s)
- Alexandria M Szalanczy
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| | - Gina Giorgio
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| | - Emily Goff
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| | - Osborne Seshie
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| | - Michael Grzybowski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jason Klotz
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eva E Redei
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leah C Solberg Woods
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| |
Collapse
|
17
|
Herselman MF, Lin L, Luo S, Yamanaka A, Zhou XF, Bobrovskaya L. Sex-Dependent Effects of Chronic Restraint Stress on Mood-Related Behaviours and Neurochemistry in Mice. Int J Mol Sci 2023; 24:10353. [PMID: 37373499 DOI: 10.3390/ijms241210353] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Anxiety and depressive disorders are closely associated; however, the pathophysiology of these disorders remains poorly understood. Further exploration of the mechanisms involved in anxiety and depression such as the stress response may provide new knowledge that will contribute to our understanding of these disorders. Fifty-eight 8-12-week-old C57BL6 mice were separated into experimental groups by sex as follows: male controls (n = 14), male restraint stress (n = 14), female controls (n = 15) and female restraint stress (n = 15). These mice were taken through a 4-week randomised chronic restraint stress protocol, and their behaviour, as well as tryptophan metabolism and synaptic proteins, were measured in the prefrontal cortex and hippocampus. Adrenal catecholamine regulation was also measured. The female mice showed greater anxiety-like behaviour than their male counterparts. Tryptophan metabolism was unaffected by stress, but some basal sex characteristics were noted. Synaptic proteins were reduced in the hippocampus in stressed females but increased in the prefrontal cortex of all female mice. These changes were not found in any males. Finally, the stressed female mice showed increased catecholamine biosynthesis capability, but this effect was not found in males. Future studies in animal models should consider these sex differences when evaluating mechanisms related to chronic stress and depression.
Collapse
Affiliation(s)
- Mauritz Frederick Herselman
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Liying Lin
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Shayan Luo
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | | | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| |
Collapse
|
18
|
Idrissi SE, Fath N, Ibork H, Taghzouti K, Alamy M, Abboussi O. Restraint Stress Exacerbates Apoptosis in a 6-OHDA Animal Model of Parkinson Disease. Neurotox Res 2023; 41:166-176. [PMID: 36633788 DOI: 10.1007/s12640-022-00630-3] [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: 10/23/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023]
Abstract
Activation of the apoptotic pathway has been associated with promoting neuronal cell death in the pathophysiology of Parkinson disease (PD). Nonetheless, the mechanisms by which it may occur remain unclear. It has been suggested that stress-induced oxidation and potential apoptosis may play a major role in the progression of PD. Thus, in this study, we aimed to investigate the effect of subchronic restraint stress on striatal dopaminergic activity, iron, p53, caspase-3, and plasmatic acetylcholinesterase (AChE) levels in male Wistar rat model of PD induced by administration of 6-hydroxydopamine (6-OHDA) in the medial forebrain bundle (MFB). The obtained results showed that restraint stress exacerbates motor coordination deficits and anxiety in animals treated with 6-OHDA in comparison to animals receiving saline, and it had no effect on object recognition memory. On another hand, 6-OHDA decreased dopamine (DA) levels, increased iron accumulation, and induced overexpression of the pro-apoptotic factors caspase-3, p53, and AChE. More interestingly, post-lesion restraint stress exacerbated the expression of caspase-3 and AChE without affecting p53 expression. These findings suggest that subchronic stress may accentuate apoptosis and may contribute to DA neuronal loss in the striatal regions and possibly exacerbate the progression of PD.
Collapse
Affiliation(s)
- Sara El Idrissi
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Nada Fath
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Hind Ibork
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Khalid Taghzouti
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Meryem Alamy
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Oualid Abboussi
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco.
| |
Collapse
|
19
|
Zhu W, Li W, Jiang J, Wang D, Mao X, Zhang J, Zhang X, Chang J, Yao P, Yang X, Da Costa C, Zhang Y, Yu J, Li H, Li S, Chi X, Li N. Chronic salmon calcitonin exerts an antidepressant effect via modulating the p38 MAPK signaling pathway. Front Mol Neurosci 2023; 16:1071327. [PMID: 36969556 PMCID: PMC10036804 DOI: 10.3389/fnmol.2023.1071327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/15/2023] [Indexed: 03/12/2023] Open
Abstract
Depression is a common recurrent psychiatric disorder with a high lifetime prevalence and suicide rate. At present, although several traditional clinical drugs such as fluoxetine and ketamine, are widely used, medications with a high efficiency and reduced side effects are of urgent need. Our group has recently reported that a single administration of salmon calcitonin (sCT) could ameliorate a depressive-like phenotype via the amylin signaling pathway in a mouse model established by chronic restraint stress (CRS). However, the molecular mechanism underlying the antidepressant effect needs to be addressed. In this study, we investigated the antidepressant potential of sCT applied chronically and its underlying mechanism. In addition, using transcriptomics, we found the MAPK signaling pathway was upregulated in the hippocampus of CRS-treated mice. Further phosphorylation levels of ERK/p38/JNK kinases were also enhanced, and sCT treatment was able only to downregulate the phosphorylation level of p38/JNK, with phosphorylated ERK level unaffected. Finally, we found that the antidepressant effect of sCT was blocked by p38 agonists rather than JNK agonists. These results provide a mechanistic explanation of the antidepressant effect of sCT, suggesting its potential for treating the depressive disorder in the clinic.
Collapse
Affiliation(s)
- Wenhui Zhu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weifen Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jian Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dilong Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xinliang Mao
- Perfect Life and Health Institute, Zhongshan, Guangdong, China
| | - Jin Zhang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xunzhi Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Jinlong Chang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Peijia Yao
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiuyan Yang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | | | - Ying Zhang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiezhong Yu
- The Fourth People’s Hospital of Datong City, Datong, China
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, Division of Medicine, Faculty of Medical Sciences, University College London, London, United Kingdom
- China-UK Institute for Frontier Science, Shenzhen, China
- *Correspondence: Huiliang Li,
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- Shupeng Li,
| | - Xinjin Chi
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Xinjin Chi,
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- China-UK Institute for Frontier Science, Shenzhen, China
- The Fifth People’s Hospital of Datong City, Datong, China
- Ningning Li,
| |
Collapse
|
20
|
Huang Y, Wang J, Liu F, Wang C, Xiao Z, Zhou W. Liuwei Dihuang formula ameliorates chronic stress-induced emotional and cognitive impairments in mice by elevating hippocampal O-GlcNAc modification. Front Neurosci 2023; 17:1134176. [PMID: 37152609 PMCID: PMC10157057 DOI: 10.3389/fnins.2023.1134176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/28/2023] [Indexed: 05/09/2023] Open
Abstract
A substantial body of evidence has indicated that intracerebral O-linked N-acetyl-β-D-glucosamine (O-GlcNAc), a generalized post-translational modification, was emerging as an effective regulator of stress-induced emotional and cognitive impairments. Our previous studies showed that the Liuwei Dihuang formula (LW) significantly improved the emotional and cognitive dysfunctions in various types of stress mouse models. In the current study, we sought to determine the effects of LW on intracerebral O-GlcNAc levels in chronic unpredictable mild stress (CUMS) mice. The dynamic behavioral tests showed that anxiety- and depression-like behaviors and object recognition memory of CUMS mice were improved in a dose-dependent manner after LW treatment. Moreover, linear discriminate analysis (LEfSe) of genera abundance revealed a significant difference in microbiome among the study groups. LW showed a great impact on the relative abundance of these gut microbiota in CUMS mice and reinstated them to control mouse levels. We found that LW potentially altered the Uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) biosynthesis process, and the abundance of O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT) in CUMS mice, which was inferred using PICRUSt analysis. We further verified advantageous changes in hippocampal O-GlcNAc modification of CUMS mice following LW administration, as well as changes in the levels of OGA and OGT. In summary, LW intervention increased the levels of hippocampal O-GlcNAc modification and ameliorated the emotional and cognitive impairments induced by chronic stress in CUMS mice. LW therefore could be considered a potential prophylactic and therapeutic agent for chronic stress.
Collapse
Affiliation(s)
- Yan Huang
- Nanjing University of Chinese Medicine, Nanjing, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Jianhui Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Feng Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Chenran Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
- *Correspondence: Zhiyong Xiao,
| | - Wenxia Zhou
- Nanjing University of Chinese Medicine, Nanjing, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
- Wenxia Zhou,
| |
Collapse
|
21
|
Zhang YD, Shi DD, Zhang S, Wang Z. Sex-specific transcriptional signatures in the medial prefrontal cortex underlying sexually dimorphic behavioural responses to stress in rats. J Psychiatry Neurosci 2023; 48:E61-E73. [PMID: 36796857 PMCID: PMC9943549 DOI: 10.1503/jpn.220147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/03/2022] [Accepted: 11/18/2022] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Converging evidence suggests that stress alters behavioural responses in a sex-specific manner; however, the underlying molecular mechanisms of stress remain largely unknown. METHODS We adapted unpredictable maternal separation (UMS) and adult restraint stress (RS) paradigms to mimic stress in rats in early life or adulthood, respectively. The sexual dimorphism of the prefrontal cortex was noted, and we performed RNA sequencing (RNA-Seq) to identify specific genes or pathways responsible for sexually dimorphic responses to stress. We then performed quantitative reverse transcription polymerase chain reaction (qRT-PCR) to verify the results of RNA-Seq. RESULTS Female rats exposed to either UMS or RS showed no negative effects on anxiety-like behaviours, whereas the emotional functions of the PFC were impaired markedly in stressed male rats. Leveraging differentially expressed genes (DEG) analyses, we identified sex-specific transcriptional profiles associated with stress. There were many overlapping DEGs between UMS and RS transcriptional data sets, where 1406 DEGs were associated with both biological sex and stress, while only 117 DEGs were related to stress. Notably, Uba52 and Rpl34-ps1 were the first-ranked hub gene in 1406 and 117 DEGs respectively, and Uba52 was higher than Rp134-ps1, suggesting that stress may have led to a more pronounced effect on the set of 1406 DEGs. Pathway analysis revealed that 1406 DEGs were primarily enriched in ribosomal pathway. These results were confirmed by qRT-PCR. LIMITATIONS Sex-specific transcriptional profiles associated with stress were identified in this study, but more in-depth experiments, such as single-cell sequencing and manipulation of male and female gene networks in vivo, are needed to verify our findings. CONCLUSION Our findings show sex-specific behavioural responses to stress and highlight sexual dimorphism at the transcriptional level, shedding light on developing sex-specific therapeutic strategies for stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Ying-Dan Zhang
- From the Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Y.-D. Zhang, Shi, S. Zhang, Wang); the Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Shi, S. Zhang, Wang); and the Institute of Psychological and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China (Wang)
| | - Dong-Dong Shi
- From the Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Y.-D. Zhang, Shi, S. Zhang, Wang); the Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Shi, S. Zhang, Wang); and the Institute of Psychological and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China (Wang)
| | - Sen Zhang
- From the Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Y.-D. Zhang, Shi, S. Zhang, Wang); the Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Shi, S. Zhang, Wang); and the Institute of Psychological and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China (Wang)
| | - Zhen Wang
- From the Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Y.-D. Zhang, Shi, S. Zhang, Wang); the Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Shi, S. Zhang, Wang); and the Institute of Psychological and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China (Wang)
| |
Collapse
|