1
|
Su C, Huang T, Zhang M, Zhang Y, Zeng Y, Chen X. Glucocorticoid receptor signaling in the brain and its involvement in cognitive function. Neural Regen Res 2025; 20:2520-2537. [PMID: 39248182 PMCID: PMC11801288 DOI: 10.4103/nrr.nrr-d-24-00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-adrenal axis regulates the secretion of glucocorticoids in response to environmental challenges. In the brain, a nuclear receptor transcription factor, the glucocorticoid receptor, is an important component of the hypothalamic-pituitary-adrenal axis's negative feedback loop and plays a key role in regulating cognitive equilibrium and neuroplasticity. The glucocorticoid receptor influences cognitive processes, including glutamate neurotransmission, calcium signaling, and the activation of brain-derived neurotrophic factor-mediated pathways, through a combination of genomic and non-genomic mechanisms. Protein interactions within the central nervous system can alter the expression and activity of the glucocorticoid receptor, thereby affecting the hypothalamic-pituitary-adrenal axis and stress-related cognitive functions. An appropriate level of glucocorticoid receptor expression can improve cognitive function, while excessive glucocorticoid receptors or long-term exposure to glucocorticoids may lead to cognitive impairment. Patients with cognitive impairment-associated diseases, such as Alzheimer's disease, aging, depression, Parkinson's disease, Huntington's disease, stroke, and addiction, often present with dysregulation of the hypothalamic-pituitary-adrenal axis and glucocorticoid receptor expression. This review provides a comprehensive overview of the functions of the glucocorticoid receptor in the hypothalamic-pituitary-adrenal axis and cognitive activities. It emphasizes that appropriate glucocorticoid receptor signaling facilitates learning and memory, while its dysregulation can lead to cognitive impairment. This provides clues about how glucocorticoid receptor signaling can be targeted to overcome cognitive disability-related disorders.
Collapse
Affiliation(s)
- Chonglin Su
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Taiqi Huang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Meiyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Xingxing Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Shiraki H, Segi-Nishida E, Suzuki K. Effect of chronic corticosterone administration on acute stress-mediated gene expression in the cortex and hippocampus of male mice. Biochem Biophys Res Commun 2025; 762:151729. [PMID: 40199127 DOI: 10.1016/j.bbrc.2025.151729] [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: 12/13/2024] [Revised: 03/12/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Corticosterone plays an important role in the stress response, physiological regulation, and development of stress-related psychiatric disorders. Although several studies have demonstrated that chronic corticosterone induces anxiety- or depressive-related behaviors in mice, it remains unclear whether chronic corticosterone administration affects gene expression in the brain during the stress response. This study investigated whether chronic corticosterone administration has a significant effect on stress-related gene expression in the brain. Therefore, mice were chronically treated with corticosterone in drinking water and gene expression was analyzed by quantitative PCR (qPCR). Moreover, restraint stress was acutely applied as a novel stressor in mice chronically treated with corticosterone in the cortex and hippocampus. We initially found that chronic corticosterone administration altered glucocorticoid signaling-mediated gene expression, such as FK506 binding protein 5 (Fkbp5) and glucocorticoid-inducible kinase 1 (Sgk1), in the cortex and hippocampus of mice. Next, we found that restraint stress exposure elevated Fkbp5 expression in the vehicle group; however, chronic corticosterone administration occluded further induction of Fkbp5 expression after restraint stress exposure. In addition, pro-inflammatory cytokines tumor necrosis factor α (Tnfa) and interleukin-1β (Il1b) mRNA expression in the cortex and hippocampus were remarkably enhanced by restraint stress in corticosterone-treated mice, but not in the vehicle group. Collectively, our results demonstrated that chronic corticosterone administration modulates glucocorticoid signaling and uncovered the robust induction of pro-inflammatory cytokines after restraint stress exposure in chronically corticosterone-treated mice. These mechanisms may be involved in the molecular basis for the onset of stress-related mental illnesses.
Collapse
Affiliation(s)
- Hirono Shiraki
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
| | - Eri Segi-Nishida
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan.
| | - Kanzo Suzuki
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan.
| |
Collapse
|
3
|
Abouseta N, Gomaa N, Tassi A, Dixon SJ, Singh K, Pani SC. Profiling mRNA encoding glucocorticoid receptor α in saliva: Relationship to hair cortisol levels in individuals aged 15-25 years. Arch Oral Biol 2025; 171:106158. [PMID: 39672058 DOI: 10.1016/j.archoralbio.2024.106158] [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: 06/29/2024] [Revised: 11/17/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE We assessed levels of mRNA encoding two glucocorticoid receptor (GR) isoforms (GRα and GRβ) in saliva and examined their relationship with hair cortisol levels and dental caries experience. DESIGN Adolescents and young adults were assessed for dental caries experience, and hair cortisol was measured by ELISA. RNA was extracted from whole saliva using TRIzol, followed by quantitative real-time PCR analysis of GRα, GRβ, and glyceraldehyde 3-phosphate dehydrogenase (GAPDH). RESULTS GRβ mRNA was not detectable in most samples, whereas GRα mRNA was observed in all samples. There were significantly lower levels of GRα mRNA in individuals with elevated hair cortisol levels than in those with normal cortisol levels. Levels of GRα mRNA did not differ significantly in individuals with dental caries experience compared to individuals with no caries experience. CONCLUSIONS We identified and quantified mRNA encoding GRα in saliva. Its levels were inversely associated with hair cortisol (a marker of chronic stress). Although caries experience was associated with hair cortisol levels, there was no significant association between GRα levels and caries experience. Chronic stress has been proposed to be associated with reduced expression of GRα and this association appears to hold for GRα mRNA levels in saliva.
Collapse
Affiliation(s)
- Naima Abouseta
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Noha Gomaa
- Children's Health Research Institute, Lawson Health Research Institute, London, ON, Canada; Oral Diagnostic Sciences, and Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, University of Western Ontario, Lawson Health Research Institute, London, ON, Canada.
| | - Ali Tassi
- Graduate Orthodontics and Dentofacial Orthopaedics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - S Jeffrey Dixon
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Krishna Singh
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Sharat C Pani
- Schulich Dentistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| |
Collapse
|
4
|
Segi-Nishida E, Suzuki K. Regulation of adult-born and mature neurons in stress response and antidepressant action in the dentate gyrus of the hippocampus. Neurosci Res 2025; 211:10-15. [PMID: 36030966 DOI: 10.1016/j.neures.2022.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022]
Abstract
The dentate gyrus (DG) of the hippocampus has been implicated in the regulation of stress responses, and in the pathophysiology and treatment of depression. This review discusses the cellular changes caused by chronic stress and the cellular role of the DG in stress-induced behavioral changes and its antidepressant-like effects. Regarding adult-born neurogenic processes in the DG, chronic stress, such as repeated social defeat, suppresses cell proliferation during and immediately after stress; however, this effect is transient. The subsequent differentiation and survival processes are differentially regulated depending on the timing and sensitivity of stress. The activation of young adult-born neurons during stress contributes to stress resilience, while the transient increase in the survival of adult-born neurons after the cessation of stress seems to promote stress susceptibility. In mature granule neurons, the predominant cells in the DG, synaptic plasticity is suppressed by chronic stress. However, a group of mature granule neurons is activated by chronic stress. Chronic antidepressant treatment can transform mature granule neurons to a phenotype resembling that of immature neurons, characterized as "dematuration". Adult-born neurons suppress the activation of mature granule neurons during stress, indicating that local neural interactions within the DG are important for the stress response. Elucidating the stress-associated context- and timing-dependent cellular changes and functions in the DG will provide insights into stress-related psychiatric diseases.
Collapse
Affiliation(s)
- Eri Segi-Nishida
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, Japan.
| | - Kanzo Suzuki
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, Japan
| |
Collapse
|
5
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [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: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
6
|
Mazurie Z, Branchereau P, Cattaert D, Henkous N, Savona-Baron C, Vouimba RM. Acute stress differently modulates interneurons excitability and synaptic plasticity in the primary motor cortex of wild-type and SOD1 G93A mouse model of ALS. J Physiol 2024; 602:4987-5015. [PMID: 39216080 DOI: 10.1113/jp285210] [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: 06/28/2023] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Primary motor cortex (M1) network stability depends on activity of inhibitory interneurons, for which susceptibility to stress was previously demonstrated in limbic regions. Hyperexcitability in M1 following changes in the excitatory/inhibitory balance is a key pathological hallmark of amyotrophic lateral sclerosis (ALS). Using electrophysiological approaches, we assessed the impact of acute restraint stress on inhibitory interneurons excitability and global synaptic plasticity in M1 of the SOD1G93A ALS mouse model at a late pre-symptomatic stage (10-12.5 weeks). Based on their firing type (continuous, discontinuous, with accommodation or not) and electrophysiological characteristics (resting potential, rheobase, firing frequency), interneurons from M1 slices were separated into four clusters, labelled from 1 to 4. Among them, only interneurons from the first cluster, presenting continuous firing with few accommodations, tended to show increased excitability in wild-type (WT) and decreased excitability in SOD1G93A animals following stress. In vivo analyses of evoked field potentials showed that stress suppressed the theta burst-induced plasticity of an excitatory component (N1) recorded in the superficial layers of M1 in WT, with no impact on an inhibitory complex (N2-P1) from the deeper layers. In SOD1G93A mice, stress did not affect N1 but suppressed the N2-P1 plasticity. These data suggest that stress can alter M1 network functioning in a different manner in WT and SOD1G93A mice, possibly through changes of inhibitory interneurons excitability and synaptic plasticity. This suggests that stress-induced activity changes in M1 may therefore influence ALS outcomes. KEY POINTS: Disruption of the excitatory/inhibitory balance in the primary motor cortex (M1) has been linked to cortical hyperexcitability development, a key pathological hallmark of amyotrophic lateral sclerosis (ALS). Psychological stress was reported to influence excitatory/inhibitory balance in limbic regions, but very little is known about its influence on the M1 functioning under physiological or pathological conditions. Our study revealed that acute stress influences the excitatory/inhibitory balance within the M1, through changes in interneurons excitability along with network plasticity. Such changes were different in pathological (SOD1G93A ALS mouse model) vs. physiological (wild-type) conditions. The results of our study help us to better understand how stress modulates the M1 and highlight the need to further characterize stress-induced motor cortex changes because it may be of importance when evaluating ALS outcomes.
Collapse
Affiliation(s)
- Zoé Mazurie
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Pascal Branchereau
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Daniel Cattaert
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Nadia Henkous
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Catherine Savona-Baron
- Present address: BoRdeaux Institute of onCology (BRIC), INSERM U1312, University of Bordeaux, Bordeaux, France
| | - Rose-Marie Vouimba
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), CNRS, UMR 5287, University of Bordeaux, Bordeaux, France
| |
Collapse
|
7
|
Lalonde R, Strazielle C. Neurochemical Anatomy of Cushing's Syndrome. Neurochem Res 2024; 49:1945-1964. [PMID: 38833089 DOI: 10.1007/s11064-024-04172-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/05/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
The neurochemical anatomy underlying Cushing's syndrome is examined for regional brain metabolism as well as neurotransmitter levels and receptor binding of biogenic amines and amino acids. Preliminary studies generally indicate that glucose uptake, blood flow, and activation on fMRI scans decreased in neocortical areas and increased in subcortical areas of patients with Cushing's syndrome or disease. Glucocorticoid-mediated increases in hippocampal metabolism occurred despite in vitro evidence of glucocorticoid-induced decreases in glucose uptake or consumption, indicating that in vivo increases are the result of indirect, compensatory, or preliminary responses. In animal studies, glucocorticoid administration decreased 5HT levels and 5HT1A receptor binding in several brain regions while adrenalectomy increased such binding. Region-specific effects were also obtained in regard to the dopaminergic system, with predominant actions of glucocorticoid-induced potentiation of reuptake blockers and releasing agents. More in-depth neuroanatomical analyses are warranted of these and amino acid-related neurotransmission.
Collapse
Affiliation(s)
- Robert Lalonde
- Laboratory of Stress, Immunity, Pathogens (UR SIMPA), University of Lorraine, Campus Santé, Bât A/B 9, avenue de la Forêt de Haye, Vandoeuvre-les-Nancy, 54500, France.
| | - Catherine Strazielle
- Laboratory of Stress, Immunity, Pathogens (UR SIMPA), University of Lorraine, Campus Santé, Bât A/B 9, avenue de la Forêt de Haye, Vandoeuvre-les-Nancy, 54500, France
- CHRU Nancy, Vandoeuvre-les-Nancy, France
| |
Collapse
|
8
|
Choudhary D, Sasibhushana RB, Shankaranarayana Rao BS, Srikumar BN. Mifepristone blocks the anxiolytic- and antidepressant-like effects of allopregnanolone in male rats. Int J Neurosci 2024; 134:839-848. [PMID: 36469636 DOI: 10.1080/00207454.2022.2153047] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Allopregnanolone (3α, 5α-tetrahydroprogesterone) is an inhibitory neurosteroid synthesized from progesterone via 5α-reductase activity in the brain and has anxiolytic, antidepressant, sedative, anticonvulsant, and analgesic activity. Altered levels of allopregnanolone cause anxiety, depression, premenstrual syndrome, and psychiatric disorders. Although allopregnanolone exerts most of its actions by modulating GABAA receptor, NMDA receptor, BDNF expression, and PXR activity, a recent study showed its effects are blocked by mifepristone on lordosis behavior which indicates the involvement of progestin or glucocorticoid receptors in the effects of allopregnanolone since mifepristone blocks both these receptors. However, whether these receptors are involved in acute anxiolytic or antidepressant-like effects is unknown. METHODS Adult male Wistar rats were used to study whether the prior administration of mifepristone would alter the effects of allopregnanolone in the elevated plus maze (EPM) and forced swim test (FST) was evaluated. RESULTS 10 mg/Kg dose of allopregnanolone increased percent open arm entries in the EPM, whereas 3 mg/Kg dose of allopregnanolone decreased percent immobility in the FST. Mifepristone administration resulted in a U-shaped response in the FST (with 1 mg/Kg, s.c., decreasing the immobility time) without significantly impacting the behavior in the EPM. In combination studies, mifepristone blocked the anxiolytic and antidepressant effects of allopregnanolone. CONCLUSION The current study provides evidence for the first time that progestin or glucocorticoid receptors are involved in the acute anxiolytic and antidepressant effects of allopregnanolone. Understanding the mechanism of action of allopregnanolone will help us design better therapeutic strategies to treat neuropsychiatric diseases such as depression and anxiety.
Collapse
Affiliation(s)
- Divya Choudhary
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
- Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, India
| | - R B Sasibhushana
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - B S Shankaranarayana Rao
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - B N Srikumar
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| |
Collapse
|
9
|
Mandolfo O, Parker H, Aguado È, Ishikawa Learmonth Y, Liao AY, O'Leary C, Ellison S, Forte G, Taylor J, Wood S, Searle R, Holley RJ, Boutin H, Bigger BW. Systemic immune challenge exacerbates neurodegeneration in a model of neurological lysosomal disease. EMBO Mol Med 2024; 16:1579-1602. [PMID: 38890537 PMCID: PMC11251277 DOI: 10.1038/s44321-024-00092-4] [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: 07/07/2023] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA) is a rare paediatric lysosomal storage disorder, caused by the progressive accumulation of heparan sulphate, resulting in neurocognitive decline and behavioural abnormalities. Anecdotal reports from paediatricians indicate a more severe neurodegeneration in MPS IIIA patients, following infection, suggesting inflammation as a potential driver of neuropathology. To test this hypothesis, we performed acute studies in which WT and MPS IIIA mice were challenged with the TLR3-dependent viral mimetic poly(I:C). The challenge with an acute high poly(I:C) dose exacerbated systemic and brain cytokine expression, especially IL-1β in the hippocampus. This was accompanied by an increase in caspase-1 activity within the brain of MPS IIIA mice with concomitant loss of hippocampal GFAP and NeuN expression. Similar levels of cell damage, together with exacerbation of gliosis, were also observed in MPS IIIA mice following low chronic poly(I:C) dosing. While further investigation is warranted to fully understand the extent of IL-1β involvement in MPS IIIA exacerbated neurodegeneration, our data robustly reinforces our previous findings, indicating IL-1β as a pivotal catalyst for neuropathological processes in MPS IIIA.
Collapse
Affiliation(s)
- Oriana Mandolfo
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Helen Parker
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Èlia Aguado
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Yuko Ishikawa Learmonth
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Ai Yin Liao
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Claire O'Leary
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Stuart Ellison
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Gabriella Forte
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Jessica Taylor
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Shaun Wood
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Rachel Searle
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Rebecca J Holley
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Hervé Boutin
- Division of Neuroscience & Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- INSERM, UMR 1253, iBrain, Université de Tours, Tours, France
| | - Brian W Bigger
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK.
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
10
|
Sherman BE, Huang I, Wijaya EG, Turk-Browne NB, Goldfarb EV. Acute Stress Effects on Statistical Learning and Episodic Memory. J Cogn Neurosci 2024; 36:1741-1759. [PMID: 38713878 PMCID: PMC11223726 DOI: 10.1162/jocn_a_02178] [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] [Indexed: 05/09/2024]
Abstract
Stress is widely considered to negatively impact hippocampal function, thus impairing episodic memory. However, the hippocampus is not merely the seat of episodic memory. Rather, it also (via distinct circuitry) supports statistical learning. On the basis of rodent work suggesting that stress may impair the hippocampal pathway involved in episodic memory while sparing or enhancing the pathway involved in statistical learning, we developed a behavioral experiment to investigate the effects of acute stress on both episodic memory and statistical learning in humans. Participants were randomly assigned to one of three conditions: stress (socially evaluated cold pressor) immediately before learning, stress ∼15 min before learning, or no stress. In the learning task, participants viewed a series of trial-unique scenes (allowing for episodic encoding of each image) in which certain scene categories reliably followed one another (allowing for statistical learning of associations between paired categories). Memory was assessed 24 hr later to isolate stress effects on encoding/learning rather than retrieval. We found modest support for our hypothesis that acute stress can amplify statistical learning: Only participants stressed ∼15 min in advance exhibited reliable evidence of learning across multiple measures. Furthermore, stress-induced cortisol levels predicted statistical learning retention 24 hr later. In contrast, episodic memory did not differ by stress condition, although we did find preliminary evidence that acute stress promoted memory for statistically predictable information and attenuated competition between statistical and episodic encoding. Together, these findings provide initial insights into how stress may differentially modulate learning processes within the hippocampus.
Collapse
|
11
|
Zheng B, Zheng Y, Hu W, Chen Z. Dissecting the networks underlying diverse brain disorders after prenatal glucocorticoid overexposure. Arch Toxicol 2024; 98:1975-1990. [PMID: 38581585 DOI: 10.1007/s00204-024-03733-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/07/2024] [Indexed: 04/08/2024]
Abstract
New human life begins in the uterus in a period of both extreme plasticity and sensitivity to environmental disturbances. The fetal stage is also a vital period for central nervous system development, with experiences at this point profoundly and permanently shaping brain structure and function. As such, some brain disorders may originate in utero. Glucocorticoids, a class of essential stress hormones, play indispensable roles in fetal development, but overexposure may have lasting impacts on the brain. In this review, we summarize data from recent clinical and non-clinical studies regarding alterations in fetal brains due to prenatal glucocorticoid overexposure that are associated with nervous system disorders. We discuss relevant changes to brain structure and cellular functions and explore the underlying molecular mechanisms. In addition, we summarize factors that may cause differential outcomes between varying brain regions, and outline clinically feasible intervention strategies that are expected to minimize negative consequences arising from fetal glucocorticoid overexposure. Finally, we highlight the need for experimental evidence aided by new technologies to clearly determine the effects of excessive prenatal glucocorticoid exposure. This review consolidates diverse findings to help researchers better understand the relationship between the prenatal glucocorticoid overexposure and the effects it has on various fetal brain regions, promoting further development of critical intervention strategies.
Collapse
Affiliation(s)
- Baixiu Zheng
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Weiwei Hu
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Zhong Chen
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
12
|
Stark R. The olfactory bulb: A neuroendocrine spotlight on feeding and metabolism. J Neuroendocrinol 2024; 36:e13382. [PMID: 38468186 DOI: 10.1111/jne.13382] [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: 11/23/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/13/2024]
Abstract
Olfaction is the most ancient sense and is needed for food-seeking, danger protection, mating and survival. It is often the first sensory modality to perceive changes in the external environment, before sight, taste or sound. Odour molecules activate olfactory sensory neurons that reside on the olfactory epithelium in the nasal cavity, which transmits this odour-specific information to the olfactory bulb (OB), where it is relayed to higher brain regions involved in olfactory perception and behaviour. Besides odour processing, recent studies suggest that the OB extends its function into the regulation of food intake and energy balance. Furthermore, numerous hormone receptors associated with appetite and metabolism are expressed within the OB, suggesting a neuroendocrine role outside the hypothalamus. Olfactory cues are important to promote food preparatory behaviours and consumption, such as enhancing appetite and salivation. In addition, altered metabolism or energy state (fasting, satiety and overnutrition) can change olfactory processing and perception. Similarly, various animal models and human pathologies indicate a strong link between olfactory impairment and metabolic dysfunction. Therefore, understanding the nature of this reciprocal relationship is critical to understand how olfactory or metabolic disorders arise. This present review elaborates on the connection between olfaction, feeding behaviour and metabolism and will shed light on the neuroendocrine role of the OB as an interface between the external and internal environments. Elucidating the specific mechanisms by which olfactory signals are integrated and translated into metabolic responses holds promise for the development of targeted therapeutic strategies and interventions aimed at modulating appetite and promoting metabolic health.
Collapse
Affiliation(s)
- Romana Stark
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
13
|
Kniffin A, Bangasser DA, Parikh V. Septohippocampal cholinergic system at the intersection of stress and cognition: Current trends and translational implications. Eur J Neurosci 2024; 59:2155-2180. [PMID: 37118907 PMCID: PMC10875782 DOI: 10.1111/ejn.15999] [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: 09/27/2022] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 04/30/2023]
Abstract
Deficits in hippocampus-dependent memory processes are common across psychiatric and neurodegenerative disorders such as depression, anxiety and Alzheimer's disease. Moreover, stress is a major environmental risk factor for these pathologies and it exerts detrimental effects on hippocampal functioning via the activation of hypothalamic-pituitary-adrenal (HPA) axis. The medial septum cholinergic neurons extensively innervate the hippocampus. Although, the cholinergic septohippocampal pathway (SHP) has long been implicated in learning and memory, its involvement in mediating the adaptive and maladaptive impact of stress on mnemonic processes remains less clear. Here, we discuss current research highlighting the contributions of cholinergic SHP in modulating memory encoding, consolidation and retrieval. Then, we present evidence supporting the view that neurobiological interactions between HPA axis stress response and cholinergic signalling impact hippocampal computations. Finally, we critically discuss potential challenges and opportunities to target cholinergic SHP as a therapeutic strategy to improve cognitive impairments in stress-related disorders. We argue that such efforts should consider recent conceptualisations on the dynamic nature of cholinergic signalling in modulating distinct subcomponents of memory and its interactions with cellular substrates that regulate the adaptive stress response.
Collapse
Affiliation(s)
- Alyssa Kniffin
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA 19122
| | - Debra A. Bangasser
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA
| | - Vinay Parikh
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA 19122
| |
Collapse
|
14
|
Pegueros-Maldonado R, Pech-Pool SM, Blancas JJ, Prado-Alcalá RA, Arámburo C, Luna M, Quirarte GL. Inhibition of corticosterone synthesis impairs cued water maze consolidation, but it does not affect the expression of BDNF, CK2 and SGK1 genes in dorsal striatum. Front Behav Neurosci 2024; 18:1341883. [PMID: 38468708 PMCID: PMC10925660 DOI: 10.3389/fnbeh.2024.1341883] [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: 11/21/2023] [Accepted: 01/30/2024] [Indexed: 03/13/2024] Open
Abstract
Corticosterone (CORT) release during learning experiences is associated with strong memories and activity of the glucocorticoid receptor. It has been shown that lesions of the dorsal striatum (DS) of rats trained in the cued version of the Morris water maze impair memory, and that local injection of CORT improves its performance, suggesting that DS activity is involved in procedural memory which may be modulated by CORT. We trained rats in cued Morris water maze and analyzed the effect of CORT synthesis inhibition on performance, CORT levels, expression of plasticity-involved genes, such as the brain derived neurotrophic factor (BDNF), casein kinase 2 (CK2), and the serum/glucocorticoid regulated kinase 1 (SGK1), as well as the presence of phosphorylated nuclear glucocorticoid receptor in serine 232 (pGR-S232) in the DS. The inhibition of CORT synthesis by metyrapone reduced CORT levels in plasma, prevented its increment in DS and impaired the performance of cued water maze. Additionally, there was an increase of CK2 and SGK1 mRNAs expression in trained subjects, which was unrelated to CORT levels. Finally, we did not observe changes in nuclear pGR-S232 in any condition. Our findings agree with evidence demonstrating that decreasing CORT levels hinders acquisition and consolidation of the spatial version of the Morris water maze; these novel findings broaden our knowledge about the involvement of the DS in the mechanisms underlying procedural memory.
Collapse
Affiliation(s)
- Rogelio Pegueros-Maldonado
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Santiago M. Pech-Pool
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Jaisson J. Blancas
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Roberto A. Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Carlos Arámburo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Maricela Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Gina L. Quirarte
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| |
Collapse
|
15
|
Araki R, Kita A, Yabe T. Decreased Brain pH Underlies Behavioral and Brain Abnormalities Induced by Chronic Exposure to Glucocorticoids in Mice. Biol Pharm Bull 2024; 47:1836-1845. [PMID: 39522977 DOI: 10.1248/bpb.b24-00251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Depressed patients may exhibit glucocorticoid hypersecretion, suggesting that elevated levels of glucocorticoids may play an important role in the pathophysiology of depression. Some postmortem brain studies have shown decreased pH and increased lactate levels in psychiatric patients, implying involvement of these factors in the pathogenesis. To investigate the effects of glucocorticoids on brain pH and lactate levels, and their roles in depressive symptoms, brain pH and lactate were examined in mice treated with corticosterone (CORT), the major bioactive glucocorticoid in rodents. A single administration of CORT decreased hippocampal pH after 24 h. Three weeks of CORT treatment decreased pH in the prefrontal cortex (PFC), striatum, and hippocampus (HC), whereas intake of pH 9.0 drinking water increased pH in these brain regions. pH and lactate levels were correlated in the PFC and HC of mice treated with CORT for 3 weeks. The suppression of body weight gain and decrease in adrenal weight observed after 3 weeks of CORT treatment were not alleviated by pH 9.0 water. However, an increase in immobility time in the forced swim test and a decrease in neurogenesis in the hippocampus were alleviated. The decrease in brain pH and increase in immobility time in the forced swim test and a decrease in neurogenesis in the hippocampus induced by CORT treatment were abolished by co-treatment with the glucocorticoid receptor (GR) antagonist mifepristone. These findings indicate that decreased brain pH via GRs may be related to glucocorticoid-induced depression-like behavior and decreased hippocampal neurogenesis.
Collapse
Affiliation(s)
- Ryota Araki
- Laboratory of Functional Biomolecules and Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Ayami Kita
- Laboratory of Functional Biomolecules and Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Takeshi Yabe
- Laboratory of Functional Biomolecules and Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| |
Collapse
|
16
|
Gorthy AS, Balleste AF, Placeres-Uray F, Atkins CM. Chronic Stress in Early Development and Effects on Traumatic Brain Injury Outcome. ADVANCES IN NEUROBIOLOGY 2024; 42:179-204. [PMID: 39432043 PMCID: PMC11556197 DOI: 10.1007/978-3-031-69832-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
In recent years, significant advances have been made in the study of mild traumatic brain injury (mTBI). Complete recovery from mTBI normally requires days to weeks, yet a subset of the population suffers from symptoms for weeks to months after injury. The risk factors for these prolonged symptoms have not yet been fully understood. In this chapter, we address one proposed risk factor, early life stress (ELS) and its influence on mTBI recovery. To study the effects of ELS on mTBI recovery, accepted animal models of ELS, including maternal separation, limited bedding and nesting, and chronic unpredictable stress, have been implemented. Combining these ELS models with standardized mTBI models, such as fluid percussion injury or controlled cortical impact, has allowed for a deeper understanding of the neuronal, hormonal, and cognitive changes that occur after mTBI following ELS. These preclinical findings are being used to understand how adverse childhood experiences may predispose a subset of individuals to poorer recovery after mTBI.
Collapse
Affiliation(s)
- Aditi S Gorthy
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alyssa F Balleste
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fabiola Placeres-Uray
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Coleen M Atkins
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
17
|
Mazurka R, Cunningham S, Hassel S, Foster JA, Nogovitsyn N, Fiori LM, Strother SC, Arnott SR, Frey BN, Lam RW, MacQueen GM, Milev RV, Rotzinger S, Turecki G, Kennedy SH, Harkness KL. Relation of hippocampal volume and SGK1 gene expression to treatment remission in major depression is moderated by childhood maltreatment: A CAN-BIND-1 report. Eur Neuropsychopharmacol 2024; 78:71-80. [PMID: 38128154 DOI: 10.1016/j.euroneuro.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 12/01/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Preclinical research implicates stress-induced upregulation of the enzyme, serum- and glucocorticoid-regulated kinase 1 (SGK1), in reduced hippocampal volume. In the current study, we tested the hypothesis that greater SGK1 mRNA expression in humans would be associated with lower hippocampal volume, but only among those with a history of prolonged stress exposure, operationalized as childhood maltreatment (physical, sexual, and/or emotional abuse). Further, we examined whether baseline levels of SGK1 and hippocampal volume, or changes in these markers over the course of antidepressant treatment, would predict treatment outcomes in adults with major depression [MDD]. We assessed SGK1 mRNA expression from peripheral blood, and left and right hippocampal volume at baseline, as well as change in these markers over the first 8 weeks of a 16-week open-label trial of escitalopram as part of the Canadian Biomarker Integration Network in Depression program (MDD [n = 161] and healthy comparison participants [n = 91]). Childhood maltreatment was assessed via contextual interview with standardized ratings. In the full sample at baseline, greater SGK1 expression was associated with lower hippocampal volume, but only among those with more severe childhood maltreatment. In individuals with MDD, decreases in SGK1 expression predicted lower remission rates at week 16, again only among those with more severe maltreatment. Decreases in hippocampal volume predicted lower week 16 remission for those with low childhood maltreatment. These results suggest that both glucocorticoid-related neurobiological mechanisms of the stress response and history of childhood stress exposure may be critical to understanding differential treatment outcomes in MDD. ClinicalTrials.gov: NCT01655706 Canadian Biomarker Integration Network for Depression Study.
Collapse
Affiliation(s)
- Raegan Mazurka
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada.
| | | | - Stefanie Hassel
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Jane A Foster
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Nikita Nogovitsyn
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Centre for Depression and Suicide Studies, St. Michael's Hospital, Toronto ON, Canada
| | - Laura M Fiori
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Stephen C Strother
- Rotman Research Institute, Baycrest, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Canada
| | | | - Benicio N Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Mood Disorders Program, St. Joseph's Healthcare Hamilton, ON, Canada
| | - Raymond W Lam
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Glenda M MacQueen
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Roumen V Milev
- Departments of Psychiatry and Psychology, And Providence Care Hospital, Queen's University, Kingston, ON, Canada
| | - Susan Rotzinger
- Department of Psychiatry, University of Toronto, Canada; Centre for Depression and Suicide Studies, St. Michael's Hospital, Toronto ON, Canada
| | - Gustavo Turecki
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Sidney H Kennedy
- Department of Psychiatry, University of Toronto, Canada; Centre for Depression and Suicide Studies, St. Michael's Hospital, Toronto ON, Canada
| | - Kate L Harkness
- Department of Psychology, Queen's University, Kingston, ON, Canada
| |
Collapse
|
18
|
Ai Y, Hummel T, Nie H, Yang J, Han P. Reduced neural responses to pleasant odor stimuli after acute psychological stress is associated with cortisol reactivity. Neuroimage 2023; 284:120474. [PMID: 38008298 DOI: 10.1016/j.neuroimage.2023.120474] [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: 09/21/2023] [Revised: 11/07/2023] [Accepted: 11/23/2023] [Indexed: 11/28/2023] Open
Abstract
Acute stress alters olfactory perception. However, little is known about the neural processing of olfactory stimuli after acute stress exposure and the role of cortisol in such an effect. Here, we used an event-related olfactory fMRI paradigm to investigate brain responses to odors of different valence (unpleasant, pleasant, or neutral) in healthy young adults following an acute stress (Trier Social Stress Test, TSST) induction (N = 22) or a non-stressful resting condition (N = 22). We obtained the odor pleasantness, intensity, and familiarity ratings after the acute stress induction or resting condition. We also measured the participants' perceived stress and salivary cortisol at four time points during the procedure. We found a stress-related decrease in brain activation in response to the pleasant, but not to the neutral or unpleasant odor stimuli in the right piriform cortex extending to the right amygdala, the right orbitofrontal cortex, and the right insula. In addition, activation of clusters within the regions of interest were negatively associated with individual baseline-to-peak increase in salivary cortisol levels after stress. We also found increased functional connectivity between the right piriform cortex and the right insula after stress when the pleasant odor was presented. The strength of the connectivity was positively correlated with increased perceived stress levels immediately after stress exposure. These results provide novel evidence for the effects of acute stress in attenuating the neural processing of a pleasant olfactory stimulus. Together with previous findings, the effect of acute stress on human olfactory perception appears to depend on both the valence and the concentration (e.g., peri-threshold or suprathreshold levels) of odor stimuli.
Collapse
Affiliation(s)
- Yun Ai
- Faculty of Psychology, Southwest University, Chongqing, China; MOE Key Laboratory of Cognition and Personality, Southwest University, Chongqing, China
| | - Thomas Hummel
- Interdisciplinary Centre Smell and Taste, Department of Otorhinolaryngology, TU Dresden, Dresden, Germany
| | - Haoyu Nie
- Faculty of Psychology, Southwest University, Chongqing, China
| | - Juan Yang
- Faculty of Psychology, Southwest University, Chongqing, China; MOE Key Laboratory of Cognition and Personality, Southwest University, Chongqing, China
| | - Pengfei Han
- Faculty of Psychology, Southwest University, Chongqing, China; MOE Key Laboratory of Cognition and Personality, Southwest University, Chongqing, China.
| |
Collapse
|
19
|
Hernandez M, Ghislin S, Lalonde R, Strazielle C. Corticosterone effects on postnatal cerebellar development in mice. Neurochem Int 2023; 171:105611. [PMID: 37704081 DOI: 10.1016/j.neuint.2023.105611] [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: 12/10/2022] [Revised: 08/23/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Glucocorticoids administered early in infancy can affect the architectonic organization of brain structures, particularly those with a postnatal development and resulting in long-term deficits of neuromotor function and cognition. The present study was undertaken to study the effects of daily corticosterone (CORT) injections at a pharmacological dose from postnatal days 8-15 on cerebellar and hippocampal development in mouse pups. Gene expression status for trophic factors involved in synaptic development and function as well as measures of layer thickness associated with cytochrome oxidase labelling were analyzed in the hippocampus, hypothalamus, and specific cerebellar lobules involved in motor control. Repeated CORT injections dysregulated the HPA axis with increased Crh and Nr3c1 mRNA levels in the hypothalamus and a resulting higher serum corticosterone level. The CORT treatment altered the morphology of the hippocampus and down-regulated gene transcription for corticotropin-releasing hormone (Crh) and its type-1 receptor (Crhr1), glucocorticoid receptor (Nr3c1), and brain-derived neurotrophic factor Bdnf and its receptor Ntrk2 (neurotrophic receptor tyrosine kinase 2). Similar mRNA expression decreases were found in the cerebellum for Crhr1, Crhr2, Nr3c1, and Grid2 (glutamatergic δ2 receptor). Morphological alterations and metabolic activity variations were observed in specific cerebellar lobules involved in motor control. The paramedian lobule, normally characterized by mitotic activity in the external germinative layer during the second postnatal week, was atrophic but metabolically hyperactive in its granule cell and molecular layers. On the contrary, lobules with an earlier cell proliferation displayed neurogenesis but a hypoactivated granule cell layer, suggesting a developmental delay in synaptogenesis. The results indicate that glucocorticoid, administered daily during the second postnatal week modulated the developmental programming of the hippocampus and cerebellum. These growth and metabolic alterations may lead possibly to morphological and functional changes later in life.
Collapse
Affiliation(s)
- M Hernandez
- Laboratory of Stress, Immunity, Pathogens (EA 7300), Medical School, University of Lorraine, 54500 Vandœuvre-les-Nancy, France; CHRU Nancy, Vandœuvre-les-Nancy, France
| | - S Ghislin
- Laboratory of Stress, Immunity, Pathogens (EA 7300), Medical School, University of Lorraine, 54500 Vandœuvre-les-Nancy, France
| | - R Lalonde
- Laboratory of Stress, Immunity, Pathogens (EA 7300), Medical School, University of Lorraine, 54500 Vandœuvre-les-Nancy, France
| | - C Strazielle
- Laboratory of Stress, Immunity, Pathogens (EA 7300), Medical School, University of Lorraine, 54500 Vandœuvre-les-Nancy, France; CHRU Nancy, Vandœuvre-les-Nancy, France.
| |
Collapse
|
20
|
Yoshimura M, Flynn BP, Kershaw YM, Zhao Z, Ueta Y, Lightman SL, Conway-Campbell BL. Phase-shifting the circadian glucocorticoid profile induces disordered feeding behaviour by dysregulating hypothalamic neuropeptide gene expression. Commun Biol 2023; 6:998. [PMID: 37775688 PMCID: PMC10541449 DOI: 10.1038/s42003-023-05347-3] [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: 10/05/2021] [Accepted: 09/12/2023] [Indexed: 10/01/2023] Open
Abstract
Here we demonstrate, in rodents, how the timing of feeding behaviour becomes disordered when circulating glucocorticoid rhythms are dissociated from lighting cues; a phenomenon most commonly associated with shift-work and transmeridian travel 'jetlag'. Adrenalectomized rats are infused with physiological patterns of corticosterone modelled on the endogenous adrenal secretory profile, either in-phase or out-of-phase with lighting cues. For the in-phase group, food intake is significantly greater during the rats' active period compared to their inactive period; a feeding pattern similar to adrenal-intact control rats. In contrast, the feeding pattern of the out-of-phase group is significantly dysregulated. Consistent with a direct hypothalamic modulation of feeding behaviour, this altered timing is accompanied by dysregulated timing of anorexigenic and orexigenic neuropeptide gene expression. For Neuropeptide Y (Npy), we report a glucocorticoid-dependent direct transcriptional regulation mechanism mediated by the glucocorticoid receptor (GR). Taken together, our data highlight the adverse behavioural outcomes that can arise when two circadian systems have anti-phasic cues, in this case impacting on the glucocorticoid-regulation of a process as fundamental to health as feeding behaviour. Our findings further highlight the need for development of rational approaches in the prevention of metabolic dysfunction in circadian-disrupting activities such as transmeridian travel and shift-work.
Collapse
Affiliation(s)
- Mitsuhiro Yoshimura
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
- Department of Physiology, University of Occupational and Environmental Health, Japan 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Benjamin P Flynn
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Yvonne M Kershaw
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Zidong Zhao
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Yoichi Ueta
- Department of Physiology, University of Occupational and Environmental Health, Japan 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Stafford L Lightman
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Becky L Conway-Campbell
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| |
Collapse
|
21
|
Smith KB, Murack M, Ismail N. The sex-dependent and enduring impact of pubertal stress on health and disease. Brain Res Bull 2023; 200:110701. [PMID: 37422090 DOI: 10.1016/j.brainresbull.2023.110701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/02/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Illness is often predicated long before the manifestation of its symptoms. Exposure to stressful experiences particularly during critical periods of development, such as puberty and adolescence, can induce various physical and mental illnesses. Puberty is a critical period of maturation for neuroendocrine systems, such as the hypothalamic-pituitary-gonadal (HPG) and hypothalamic-pituitary-adrenal (HPA) axes. Exposure to adverse experiences during puberty can impede normal brain reorganizing and remodelling and result in enduring consequences on brain functioning and behaviour. Stress responsivity differs between the sexes during the pubertal period. This sex difference is partly due to differences in circulating sex hormones between males and females, impacting stress and immune responses differently. The effects of stress during puberty on physical and mental health remains under-examined. The purpose of this review is to summarize the most recent findings pertaining to age and sex differences in HPA axis, HPG axis, and immune system development, and describe how disruption in the functioning of these systems can propagate disease. Lastly, we delve into the notable neuroimmune contributions, sex differences, and the mediating role of the gut microbiome on stress and health outcomes. Understanding the enduring consequences of adverse experiences during puberty on physical and mental health will allow a greater proficiency in treating and preventing stress-related diseases early in development.
Collapse
Affiliation(s)
- Kevin B Smith
- NISE Laboratory - University of Ottawa, School of Psychology, Ottawa, Ontario, Canada
| | - Michael Murack
- NISE Laboratory - University of Ottawa, School of Psychology, Ottawa, Ontario, Canada
| | - Nafissa Ismail
- NISE Laboratory - University of Ottawa, School of Psychology, Ottawa, Ontario, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada; LIFE Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
22
|
González-Franco DA, Pegueros-Maldonado R, Cruz-Quiroz AM, Serafín N, Bello-Medina PC, Prado-Alcalá RA, Quirarte GL. Intense inhibitory avoidance training increases nuclear-phosphorylated glucocorticoid receptors in neurons of CA1 of hippocampus and ventral caudate putamen. Brain Res 2023; 1808:148316. [PMID: 36906227 DOI: 10.1016/j.brainres.2023.148316] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/09/2023] [Accepted: 03/04/2023] [Indexed: 03/11/2023]
Abstract
Corticosterone (CORT), the principal glucocorticoid in rodents, is released after stressful experiences such as training with high foot-shock intensities in the inhibitory avoidance task (IA). CORT reaches the glucocorticoid receptor (GR) located in almost all brain cells; the GR is subsequently phosphorylated at serine 232 (pGRser232). This has been reported as an indicator of ligand-dependent activation of the GR, as well as a requirement for its translocation into the nucleus for its transcription factor activity. The GR is present in the hippocampus with a high concentration in CA1 and dentate gyrus (DG), and a smaller proportion in CA3, and sparsely present in the caudate putamen (CPu); both structures are involved in memory consolidation of IA. To study the participation of CORT in IA, we quantified the ratio of pGR-positive neurons in both dorsal hippocampus (CA1, CA3 and DG) and dorsal and ventral regions of CPu of rats trained in IA, using different foot-shock intensities. Brains were dissected 60 min after training for immunodetection of pGRser232 positive cells. The results show that the groups trained with 1.0 and 2.0 mA had higher retention latencies than the 0.0 mA or 0.5 mA groups. An increase in the ratio of pGR-positive neurons was found in CA1 and ventral region of CPu only for the 2.0 mA trained group. These findings suggest that activation of GRs in CA1 and ventral CPu is involved in the consolidation of a stronger memory of IA, possibly through the modulation of gene expression.
Collapse
Affiliation(s)
- Diego A González-Franco
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla. C.P. 76230, Querétaro, México
| | - Rogelio Pegueros-Maldonado
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla. C.P. 76230, Querétaro, México
| | - América M Cruz-Quiroz
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla. C.P. 76230, Querétaro, México
| | - Norma Serafín
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla. C.P. 76230, Querétaro, México
| | - Paola C Bello-Medina
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla. C.P. 76230, Querétaro, México
| | - Roberto A Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla. C.P. 76230, Querétaro, México
| | - Gina L Quirarte
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla. C.P. 76230, Querétaro, México.
| |
Collapse
|
23
|
Natsaridis E, Perdikaris P, Fokos S, Dermon CR. Neuronal and Astroglial Localization of Glucocorticoid Receptor GRα in Adult Zebrafish Brain ( Danio rerio). Brain Sci 2023; 13:861. [PMID: 37371341 DOI: 10.3390/brainsci13060861] [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: 04/13/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Glucocorticoid receptor α (GRα), a ligand-regulated transcription factor, mainly activated by cortisol in humans and fish, mediates neural allostatic and homeostatic functions induced by different types of acute and chronic stress, and systemic inflammation. Zebrafish GRα is suggested to have multiple transcriptional effects essential for normal development and survival, similarly to mammals. While sequence alignments of human, monkey, rat, and mouse GRs have shown many GRα isoforms, we questioned the protein expression profile of GRα in the adult zebrafish (Danio rerio) brain using an alternative model for stress-related neuropsychiatric research, by means of Western blot, immunohistochemistry and double immunofluorescence. Our results identified four main GRα-like immunoreactive bands (95 kDa, 60 kDa, 45 kDa and 35 kDa), with the 95 kDa protein showing highest expression in forebrain compared to midbrain and hindbrain. GRα showed a wide distribution throughout the antero-posterior zebrafish brain axis, with the most prominent labeling within the telencephalon, preoptic, hypothalamus, midbrain, brain stem, central grey, locus coeruleus and cerebellum. Double immunofluorescence revealed that GRα is coexpressed in TH+, β2-AR+ and vGLUT+ neurons, suggesting the potential of GRα influences on adrenergic and glutamatergic transmission. Moreover, GRα was co-localized in midline astroglial cells (GFAP+) within the telencephalon, hypothalamus and hindbrain. Interestingly, GRα expression was evident in the brain regions involved in adaptive stress responses, social behavior, and sensory and motor integration, supporting the evolutionarily conserved features of glucocorticoid receptors in the zebrafish brain.
Collapse
Affiliation(s)
- Evangelos Natsaridis
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| | - Panagiotis Perdikaris
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| | - Stefanos Fokos
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| | - Catherine R Dermon
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| |
Collapse
|
24
|
Sulu C, Koca O, Icli TB, Oz A, Kargin OA, Durcan E, Sahin S, Arslan S, Turan S, Kadioglu P, Ozkaya HM. Altered thalamic volume in patients with mild autonomous cortisol secretion: a structural brain MRI study. Neuroradiology 2023; 65:1037-1051. [PMID: 37121916 DOI: 10.1007/s00234-023-03156-3] [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: 02/08/2023] [Accepted: 04/20/2023] [Indexed: 05/02/2023]
Abstract
PURPOSE To compare thalamic volume and cognitive functions of patients with mild autonomous cortisol secretion (MACS) with control subjects and patients with overt Cushing's syndrome (CS). METHODS In this cross-sectional study, volumes of regions of interest were assessed using 3 T magnetic resonance imaging and a voxel-based morphometry approach in 23 patients with MACS, 21 patients with active CS, 27 patients with CS in remission, and 21 control subjects. Cognitive functions were assessed using validated questionnaires. RESULTS Patients with MACS had smaller left thalamic (F = 3.8, p = 0.023), left posterior thalamic (F = 4.9, p = 0.01), left medial thalamic (F = 4.7, p = 0.028), and right lateral thalamic (F = 4.1, p = 0.025) volumes than control subjects. Patients with active CS also had smaller left thalamic (F = 3.8, p = 0.044), left posterior thalamic (F = 4.9, p = 0.007), left medial thalamic (F = 4.7, p = 0.006), and right lateral thalamic (F = 4.1, p = 0.042) volumes compared to controls. Patients with CS in remission had smaller left medial (F = 4.7, p = 0.030) and right lateral thalamic (F = 4.1, p = 0.028) volumes than controls. Neuropsychological tests showed no difference between the groups. CONCLUSION MACS may decrease thalamic volume.
Collapse
Affiliation(s)
- Cem Sulu
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Oguzhan Koca
- Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Tevhide Betul Icli
- Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Ahmet Oz
- Department of Radiology, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Osman Aykan Kargin
- Department of Radiology, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | | | - Serdar Sahin
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Serdar Arslan
- Department of Radiology, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Senol Turan
- Department of Psychiatry, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Pinar Kadioglu
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye
- Pituitary Center, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Hande Mefkure Ozkaya
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Türkiye.
- Pituitary Center, Istanbul University-Cerrahpasa, Istanbul, Türkiye.
| |
Collapse
|
25
|
Ai Y, Yang J, Nie H, Hummel T, Han P. Increased sensitivity to unpleasant odor following acute psychological stress. Horm Behav 2023; 150:105325. [PMID: 36805607 DOI: 10.1016/j.yhbeh.2023.105325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
Previous studies have reported increased sensitivity to malodor after acute stress in humans. However, it is unclear whether stress-related "hypersensitivity" to odors depends on odor pleasantness. Forty participants (mean age 19.13 ± 1.14 years, 21 men and 19 women) completed a stress (Trier Social Stress Test, TSST) and a control session in randomized order. Detection threshold to three odors varying in pleasantness (pleasant: β-Citronellol; neutral: 2-Heptanol; unpleasant: 4-Methylpentanoic acid), odor discrimination, odor identification, sensitivity to trigeminal odor, and suprathreshold odor perception were assessed after participants' completion of the stress or the control tasks. Salivary cortisol, subjective stress, and heart rate were assessed throughout the experiment. After TSST, participants showed an increased sensitivity for the unpleasant odor. Moreover, there were correlations between stress-related salivary cortisol and the increased sensitivity for the unpleasant odor (r = 0.32, p = 0.05) and the neutral odor (r = 0.34, p < 0.05). Besides, salivary cortisol response was correlated to the increased odor discrimination performance (Δ stress - control) (r = 0.34, p < 0.05). The post-TSST perceived stress was correlated with decreased odor identification and decreased sensitivity to the unpleasant odor. After stress, participants rated lower pleasantness for β-Citronellol than the control condition. Overall, these results suggest the impact of acute psychological stress on odor sensitivity depends on the odor valence, and that the stress-related cortisol responses may play an important role in this effect.
Collapse
Affiliation(s)
- Yun Ai
- Faculty of Psychology, Southwest University, Chongqing, China
| | - Juan Yang
- Faculty of Psychology, Southwest University, Chongqing, China; MOE Key Laboratory of Cognition and Personality, Southwest University, Chongqing, China
| | - Haoyu Nie
- Faculty of Psychology, Southwest University, Chongqing, China
| | - Thomas Hummel
- Interdisciplinary Centre Smell and Taste, Department of Otorhinolaryngology, TU Dresden, Dresden, Germany
| | - Pengfei Han
- Faculty of Psychology, Southwest University, Chongqing, China; MOE Key Laboratory of Cognition and Personality, Southwest University, Chongqing, China.
| |
Collapse
|
26
|
Manohar S, Chen GD, Li L, Liu X, Salvi R. Chronic stress induced loudness hyperacusis, sound avoidance and auditory cortex hyperactivity. Hear Res 2023; 431:108726. [PMID: 36905854 DOI: 10.1016/j.heares.2023.108726] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Hyperacusis, a debilitating loudness intolerance disorder, has been linked to chronic stress and adrenal insufficiency. To investigate the role of chronic stress, rats were chronically treated with corticosterone (CORT) stress hormone. Chronic CORT produced behavioral evidence of loudness hyperacusis, sound avoidance hyperacusis, and abnormal temporal integration of loudness. CORT treatment did not disrupt cochlear or brainstem function as reflected by normal distortion product otoacoustic emissions, compound action potentials, acoustic startle reflexex, and auditory brainstem responses. In contrast, the evoked response from the auditory cortex was enhanced up to three fold after CORT treatment. This hyperactivity was associated with a significant increase in glucocorticoid receptors in auditory cortex layers II/III and VI. Basal serum CORT levels remained normal after chronic CORT stress whereas reactive serum CORT levels evoked by acute restraint stress were blunted (reduced) after chronic CORT stress; similar changes were observed after chronic, intense noise stress. Taken together, our results show for the first time that chronic stress can induce hyperacusis and sound avoidance. A model is proposed in which chronic stress creates a subclinical state of adrenal insufficiency that establishes the necessary conditions for inducing hyperacusis.
Collapse
Affiliation(s)
- Senthilvelan Manohar
- Center for Hearing and Deafness, 137 Cary Hall, University at Buffalo, Buffalo, NY 14214, USA
| | - Guang-Di Chen
- Center for Hearing and Deafness, 137 Cary Hall, University at Buffalo, Buffalo, NY 14214, USA
| | - Li Li
- Center for Hearing and Deafness, 137 Cary Hall, University at Buffalo, Buffalo, NY 14214, USA
| | - Xiaopeng Liu
- Center for Hearing and Deafness, 137 Cary Hall, University at Buffalo, Buffalo, NY 14214, USA
| | - Richard Salvi
- Center for Hearing and Deafness, 137 Cary Hall, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
27
|
Thongrong S, Surapinit S, Promsrisuk T, Jittiwat J, Kongsui R. Pinostrobin alleviates chronic restraint stress‑induced cognitive impairment by modulating oxidative stress and the function of astrocytes in the hippocampus of rats. Biomed Rep 2023; 18:20. [PMID: 36798091 PMCID: PMC9922797 DOI: 10.3892/br.2023.1602] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Chronic stress has been recognized to induce the alterations of neuronal and glial cells in the hippocampus, and is thus implicated in cognitive dysfunction. There is increasing evidence to indicate that natural compounds capable of exerting neuroprotective and antioxidant activities, may function as potential therapeutic agents for cognitive impairment. The present study examined the neuroprotective effects of pinostrobin from Boesenbergia rotunda (L.) against chronic restraint stress (CRS)-induced cognitive impairment associated with the alterations of oxidative stress, neuronal density and glial fibrillary acidic protein (GFAP) of astrocytes in the hippocampus. For this purpose, male Wistar rats were administered once daily with pinostrobin (20 and 40 mg/kg, per os) prior to exposure to CRS (6 h/day) for 21 days. The cognitive behaviors, the concentration of malondialdehyde, and the activities of superoxide dismutase and catalase were determined. Histologically, the alterations in astrocytic GFAP and excitatory amino acid transporter 2 (EAAT2) in the hippocampus were examined. The results revealed that pinostrobin potentially attenuated cognitive impairment in the Y-maze and in novel object recognition tests, with a reduction in oxidative stress. Furthermore, pinostrobin effectively increased neuronal density, as well as the immunoreactivities of GFAP and EAAT2 in the hippocampus. Taken together, these findings indicate that treatment with pinostrobin alleviates chronic stress-induced cognitive impairment by exerting antioxidant effects, reducing neuronal cell damage, and improving the function of astrocytic GFAP and EAAT2. Thus, pinostrobin may have potential for use as a neuroprotective agent to protect against chronic stress-induced brain dysfunction and cognitive deficits.
Collapse
Affiliation(s)
- Sitthisak Thongrong
- Division of Anatomy, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand,Unit of Excellence in Translational Neurosciences Initiative, University of Phayao, Phayao 56000, Thailand
| | - Serm Surapinit
- Unit of Excellence in Translational Neurosciences Initiative, University of Phayao, Phayao 56000, Thailand,Department of Medical Technology, School of Allied Health Sciences, University of Phayao, Phayao 56000, Thailand
| | - Tichanon Promsrisuk
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Jinatta Jittiwat
- Faculty of Medicine, Mahasarakham University, Mahasarakham 44000, Thailand
| | - Ratchaniporn Kongsui
- Unit of Excellence in Translational Neurosciences Initiative, University of Phayao, Phayao 56000, Thailand,Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand,Correspondence to: Dr Ratchaniporn Kongsui, Division of Physiology, School of Medical Sciences, University of Phayao, 19 Moo 2 Phahonyothin Road, Maeka, Muang Phayao, Phayao 56000, Thailand
| |
Collapse
|
28
|
Effects of intra-hippocampal corticosterone and sleep on consolidation of memories of aversive experience in rats. Neurobiol Learn Mem 2023; 198:107721. [PMID: 36610686 DOI: 10.1016/j.nlm.2023.107721] [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: 02/12/2022] [Revised: 12/06/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
Formation and consolidation of memories for highly stressful (traumatic) events is a complex process that involves interplay between multiple memory systems and has implications for etiology and treatment of stress- and trauma-related disorders. Here we study effects of sleep/wake states and high intra-hippocampal corticosterone on consolidation of aversive contextual memories, as well as consolidation of association between auditory unpaired phasic background cues and fear response in rats. Animals were implanted with EEG and EMG electrodes for sleep assessment and cannulas for intra-hippocampal corticosterone application. They were familiarized to a "safe box" and then trained in a fear conditioning paradigm in a distinct "shock box" with a prominent unpaired phasic background auditory cue. Immediately after conditioning, animals received bilateral intra-hippocampal saline (1 μl) or corticosterone (10 ng in 1 μl) injection and were either allowed to sleep or were kept awake for a following two-hour consolidation period. Memory tests 24 h later revealed that the saline-injected animals that slept during consolidation had significantly stronger fear responses in the shock box compared to the safe box as well as increased fear response in response to the auditory cue. Lack of sleep during the consolidation period in saline injected animals led to generalization of the fear response to the safe context, while association between auditory cue and fear response was preserved. High intra-hippocampal corticosterone levels during memory consolidation led to generalization of fear response to the safe context, regardless of sleep/wake state, while enhancement of response to auditory cue was not observed. Our results show how manipulation of conditions during consolidation can lead to greatly variable memories for an aversive episode with distinct behavioral outcomes. Observed overgeneralization of fear to safe context and altered fear response to background phasic cue has implications for understanding etiology of pathological memory alternations in stress-related conditions e.g., in posttraumatic stress disorder in humans.
Collapse
|
29
|
Yao Y, Silver R. Mutual Shaping of Circadian Body-Wide Synchronization by the Suprachiasmatic Nucleus and Circulating Steroids. Front Behav Neurosci 2022; 16:877256. [PMID: 35722187 PMCID: PMC9200072 DOI: 10.3389/fnbeh.2022.877256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/11/2022] [Indexed: 11/18/2022] Open
Abstract
Background Steroids are lipid hormones that reach bodily tissues through the systemic circulation, and play a major role in reproduction, metabolism, and homeostasis. All of these functions and steroids themselves are under the regulation of the circadian timing system (CTS) and its cellular/molecular underpinnings. In health, cells throughout the body coordinate their daily activities to optimize responses to signals from the CTS and steroids. Misalignment of responses to these signals produces dysfunction and underlies many pathologies. Questions Addressed To explore relationships between the CTS and circulating steroids, we examine the brain clock located in the suprachiasmatic nucleus (SCN), the daily fluctuations in plasma steroids, the mechanisms producing regularly recurring fluctuations, and the actions of steroids on their receptors within the SCN. The goal is to understand the relationship between temporal control of steroid secretion and how rhythmic changes in steroids impact the SCN, which in turn modulate behavior and physiology. Evidence Surveyed The CTS is a multi-level organization producing recurrent feedback loops that operate on several time scales. We review the evidence showing that the CTS modulates the timing of secretions from the level of the hypothalamus to the steroidogenic gonadal and adrenal glands, and at specific sites within steroidogenic pathways. The SCN determines the timing of steroid hormones that then act on their cognate receptors within the brain clock. In addition, some compartments of the body-wide CTS are impacted by signals derived from food, stress, exercise etc. These in turn act on steroidogenesis to either align or misalign CTS oscillators. Finally this review provides a comprehensive exploration of the broad contribution of steroid receptors in the SCN and how these receptors in turn impact peripheral responses. Conclusion The hypothesis emerging from the recognition of steroid receptors in the SCN is that mutual shaping of responses occurs between the brain clock and fluctuating plasma steroid levels.
Collapse
Affiliation(s)
- Yifan Yao
- Department of Psychology, Columbia University, New York City, NY, United States
- *Correspondence: Yifan Yao,
| | - Rae Silver
- Department of Psychology, Columbia University, New York City, NY, United States
- Department of Neuroscience, Barnard College, New York City, NY, United States
- Department of Psychology, Barnard College, New York City, NY, United States
- Department of Pathology and Cell Biology, Graduate School, Columbia University Irving Medical Center, New York City, NY, United States
| |
Collapse
|
30
|
Sosa MK, Boorman DC, Keay KA. Sciatic nerve injury rebalances the hypothalamic-pituitary-adrenal axis in rats with persistent changes to their social behaviours. J Neuroendocrinol 2022; 34:e13131. [PMID: 35487591 PMCID: PMC9286784 DOI: 10.1111/jne.13131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/31/2022] [Accepted: 03/24/2022] [Indexed: 11/29/2022]
Abstract
Increased glucocorticoids characterise acute pain responses, but not the chronic pain state, suggesting specific modifications to the hypothalamic-pituitary-adrenal (HPA)-axis preventing the persistent nature of chronic pain from elevating basal glucocorticoid levels. Individuals with chronic pain mount normal HPA-axis responses to acute stressors, indicating a rebalancing of the circuits underpinning these responses. Preclinical models of chronic neuropathic pain generally recapitulate these clinical observations, but few studies have considered that the underlying neuroendocrine circuitry may be altered. Additionally, individual differences in the behavioural outcomes of these pain models, which are strikingly similar to the range of behavioural subpopulations that manifest in response to stress, threat and motivational cues, may also be reflected in divergent patterns of HPA-axis activity, which characterises these other behavioural subpopulations. We investigated the effects of sciatic nerve chronic constriction injury (CCI) on adrenocortical and hypothalamic markers of HPA-axis activity in the subpopulation of rats showing persistent changes in social interactions after CCI (Persistent Effect) and compared them with rats that do not show these changes (No Effect). Basal plasma corticosterone did not change after CCI and did not differ between groups. However, adrenocortical sensitivity to adrenocorticotropic hormone (ACTH) diverged between these groups. No Effect rats showed large increases in basal plasma ACTH with no change in adrenocortical melanocortin 2 receptor (MC2 R) expression, whereas Persistent Effect rats showed modest decreases in plasma ACTH and large increases in MC2 R expression. In the paraventricular nucleus of the hypothalamus of Persistent Effect rats, single labelling revealed significantly increased numbers of corticotropin releasing factor (CRF) +ve and glucocorticoid receptor (GR) +ve neurons. Double-labelling revealed fewer GR +ve CRF +ve neurons, suggesting a decreased hypothalamic sensitivity of CRF neurons to circulating corticosterone in Persistent Effect rats. We suggest that in addition to rebalancing the HPA-axis, the increased CRF expression in Persistent Effect rats contributes to changes in complex behaviours, and in particular social interactions.
Collapse
Affiliation(s)
- M. Karmina Sosa
- School of Medical Sciences and the Brain and Mind CentreThe University of SydneyCamperdownNew South WalesAustralia
| | - Damien C. Boorman
- School of Medical Sciences and the Brain and Mind CentreThe University of SydneyCamperdownNew South WalesAustralia
| | - Kevin A. Keay
- School of Medical Sciences and the Brain and Mind CentreThe University of SydneyCamperdownNew South WalesAustralia
| |
Collapse
|
31
|
Viho EMG, Buurstede JC, Berkhout JB, Mahfouz A, Meijer OC. Cell type specificity of glucocorticoid signaling in the adult mouse hippocampus. J Neuroendocrinol 2022; 34:e13072. [PMID: 34939259 PMCID: PMC9286676 DOI: 10.1111/jne.13072] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/14/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022]
Abstract
Glucocorticoid stress hormones are powerful modulators of brain function and can affect mood and cognitive processes. The hippocampus is a prominent glucocorticoid target and expresses both the glucocorticoid receptor (GR: Nr3c1) and the mineralocorticoid receptor (MR: Nr3c2). These nuclear steroid receptors act as ligand-dependent transcription factors. Transcriptional effects of glucocorticoids have often been deduced from bulk mRNA measurements or spatially informed individual gene expression. However, only sparse data exists allowing insights on glucocorticoid-driven gene transcription at the cell type level. Here, we used publicly available single-cell RNA sequencing data to assess the cell-type specificity of GR and MR signaling in the adult mouse hippocampus. The data confirmed that Nr3c1 and Nr3c2 expression differs across neuronal and non-neuronal cell populations. We analyzed co-expression with sex hormones receptors, transcriptional coregulators, and receptors for neurotransmitters and neuropeptides. Our results provide insights in the cellular basis of previous bulk mRNA results and allow the formulation of more defined hypotheses on the effects of glucocorticoids on hippocampal function.
Collapse
Affiliation(s)
- Eva M. G. Viho
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Jacobus C. Buurstede
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Jari B. Berkhout
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Ahmed Mahfouz
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
- Delft Bioinformatics LaboratoryDelft University of TechnologyDelftThe Netherlands
- Leiden Computational Biology CenterLeiden University Medical CenterLeidenThe Netherlands
| | - Onno C. Meijer
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
32
|
Beroukhim G, Esencan E, Seifer DB. Impact of sleep patterns upon female neuroendocrinology and reproductive outcomes: a comprehensive review. Reprod Biol Endocrinol 2022; 20:16. [PMID: 35042515 PMCID: PMC8764829 DOI: 10.1186/s12958-022-00889-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/06/2022] [Indexed: 12/17/2022] Open
Abstract
Sleep is vital to human bodily function. Growing evidence indicates that sleep deprivation, disruption, dysrhythmia, and disorders are associated with impaired reproductive function and poor clinical outcomes in women. These associations are largely mediated by molecular-genetic and hormonal pathways, which are crucial for the complex and time sensitive processes of hormone synthesis/secretion, folliculogenesis, ovulation, fertilization, implantation, and menstruation. Pathologic sleep patterns are closely linked to menstrual irregularity, polycystic ovarian syndrome, premature ovarian insufficiency, sub/infertility, and early pregnancy loss. Measures of success with assisted reproductive technology are also lower among women who engage in shift work, or experience sleep disruption or short sleep duration. Extremes of sleep duration, poor sleep quality, sleep disordered breathing, and shift work are also associated with several harmful conditions in pregnancy, including gestational diabetes and hypertensive disorders. While accumulating evidence implicates pathologic sleep patterns in impaired reproductive function and poor reproductive outcomes, additional research is needed to determine causality and propose therapeutic interventions.
Collapse
Affiliation(s)
- Gabriela Beroukhim
- Department of Obstetrics, Gynecology, and Reproductive Sciences at Yale School of Medicine, 333 Cedar St, New Haven, CT, 06510, USA.
| | - Ecem Esencan
- Department of Obstetrics, Gynecology, and Reproductive Sciences at Yale School of Medicine, 333 Cedar St, New Haven, CT, 06510, USA
| | - David B Seifer
- Department of Obstetrics, Gynecology, and Reproductive Sciences at Yale School of Medicine, 333 Cedar St, New Haven, CT, 06510, USA
| |
Collapse
|
33
|
Kong X, Ota SM, Suchecki D, Lan A, Peereboom AI, Hut RA, Meerlo P. Chronic Social Defeat Stress Shifts Peripheral Circadian Clocks in Male Mice in a Tissue-Specific and Time-of-Day Dependent Fashion. J Biol Rhythms 2022; 37:164-176. [PMID: 34994236 DOI: 10.1177/07487304211065336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Uncontrollable stress is linked to the development of many diseases, some of which are associated with disrupted daily rhythms in physiology and behavior. While available data indicate that the master circadian pacemaker in the suprachiasmatic nucleus (SCN) is unaffected by stress, accumulating evidence suggest that circadian oscillators in peripheral tissues and organs can be shifted by a variety of stressors and stress hormones. In the present study, we examined effects of acute and chronic social defeat stress in mice and addressed the question of whether effects of uncontrollable stress on peripheral clocks are tissue specific and depend on time of day of stress exposure. We used mice that carry a luciferase reporter gene fused to the circadian clock gene Period2 (PER2::LUC) to examine daily rhythms of PER2 expression in various peripheral tissues. Mice were exposed to social defeat stress in the early (ZT13-14) or late (ZT21-22) dark phase, either once (acute stress) or repeatedly on 10 consecutive days (chronic stress). One hour after the last stressor, tissue samples from liver, lung, kidney, and white adipose tissue (WAT) were collected. Social defeat stress caused a phase delay of several hours in the rhythm of PER2 expression in lung and kidney, but this delay was stronger after chronic than after acute stress. Moreover, shifts only occurred after stress in the late dark phase, not in the early dark phase. PER2 rhythms in liver and WAT were not significantly shifted by social defeat, suggesting a different response of various peripheral clocks to stress. This study indicates that uncontrollable social defeat stress is capable of shifting peripheral clocks in a time of day dependent and tissue specific manner. These shifts in peripheral clocks were smaller or absent after a single stress exposure and may therefore be the consequence of a cumulative chronic stress effect.
Collapse
Affiliation(s)
- Xiangpan Kong
- Chronobiology Unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands.,School of Medicine, Hunan Normal University, Changsha, P.R. China
| | - Simone M Ota
- Chronobiology Unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands.,Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deborah Suchecki
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Andy Lan
- Chronobiology Unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Anouk I Peereboom
- Chronobiology Unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Roelof A Hut
- Chronobiology Unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Peter Meerlo
- Chronobiology Unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
34
|
Liška K, Sládek M, Houdek P, Shrestha N, Lužná V, Ralph MR, Sumová A. High Sensitivity of the Circadian Clock in the Hippocampal Dentate Gyrus to Glucocorticoid- and GSK3-Beta-Dependent Signals. Neuroendocrinology 2022; 112:384-398. [PMID: 34111876 DOI: 10.1159/000517689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/07/2021] [Indexed: 11/19/2022]
Abstract
AIMS Circadian clocks in the hippocampus (HPC) align memory processing with appropriate time of day. Our study was aimed at ascertaining the specificity of glycogen synthase kinase 3-beta (GSK3β)- and glucocorticoid (GC)-dependent pathways in the entrainment of clocks in individual HPC regions, CA1-3, and dentate gyrus (DG). METHODS The role of GCs was addressed in vivo by comparing the effects of adrenalectomy (ADX) and subsequent dexamethasone (DEX) supplementation on clock gene expression profiles (Per1, Per2, Nr1d1, and Bmal1). In vitro the effects of DEX and the GSK3β inhibitor, CHIR-99021, were assessed from recordings of bioluminescence rhythms in HPC organotypic explants of mPER2Luc mice. RESULTS Circadian rhythms of clock gene expression in all HPC regions were abolished by ADX, and DEX injections to the rats rescued those rhythms in DG. The DEX treatment of the HPC explants significantly lengthened periods of the bioluminescence rhythms in all HPC regions with the most significant effect in DG. In contrast to DEX, CHIR-99021 significantly shortened the period of bioluminescence rhythm. Again, the effect was most significant in DG which lacks the endogenously inactivated (phosphorylated) form of GSK3β. Co-treatment of the explants with CHIR-99021 and DEX produced the CHIR-99021 response. Therefore, the GSK3β-mediated pathway had dominant effect on the clocks. CONCLUSION GSK3β- and GC-dependent pathways entrain the clock in individual HPC regions by modulating their periods in an opposite manner. The results provide novel insights into the mechanisms connecting the arousal state-relevant signals with temporal control of HPC-dependent memory and cognitive functions.
Collapse
Affiliation(s)
- Karolína Liška
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Martin Sládek
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Pavel Houdek
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Norzin Shrestha
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Vendula Lužná
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martin R Ralph
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Alena Sumová
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
35
|
Ha GE, Cheong E. Chronic Restraint Stress Decreases the Excitability of Hypothalamic POMC Neuron and Increases Food Intake. Exp Neurobiol 2021; 30:375-386. [PMID: 34983879 PMCID: PMC8752322 DOI: 10.5607/en21037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/08/2021] [Accepted: 12/11/2021] [Indexed: 11/19/2022] Open
Abstract
Stress activates the hypothalamic-pituitary-adrenal system, and induces the release of glucocorticoids, stress hormones, into circulation. Many studies have shown that stress affects feeding behavior, however, the underlying circuitry and molecular mechanisms are not fully understood. The balance between orexigenic (simulating appetite) and anorexigenic (loss of appetite) signals reciprocally modulate feeding behavior. It is suggested that proopiomelanocortin (POMC) and neuropeptide Y (NPY) neurons in the arcuate nucleus (ARC) of the hypothalamus are the first-order neurons that respond to the circulating signals of hunger and satiety. Here, we examined a chronic restraint stress model and observed an increase in food intake, which was not correlated with anhedonia. We investigated whether stress affects the properties of POMC and NPY neurons and found that chronic restraint stress reduced the excitatory inputs onto POMC neurons and increased the action potential threshold. Therefore, our study suggests that chronic stress modulates the intrinsic excitability and excitatory inputs in POMC neurons, leading to changes in feeding behavior.
Collapse
Affiliation(s)
- Go Eun Ha
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
36
|
Hodges TE, Puri TA, Blankers SA, Qiu W, Galea LAM. Steroid hormones and hippocampal neurogenesis in the adult mammalian brain. VITAMINS AND HORMONES 2021; 118:129-170. [PMID: 35180925 DOI: 10.1016/bs.vh.2021.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hippocampal neurogenesis persists across the lifespan in many species, including rodents and humans, and is associated with cognitive performance and the pathogenesis of neurodegenerative disease and psychiatric disorders. Neurogenesis is modulated by steroid hormones that change across development and differ between the sexes in rodents and humans. Here, we discuss the effects of stress and glucocorticoid exposure from gestation to adulthood as well as the effects of androgens and estrogens in adulthood on neurogenesis in the hippocampus. Throughout the review we highlight sex differences in the effects of steroid hormones on neurogenesis and how they may relate to hippocampal function and disease. These data highlight the importance of examining age and sex when evaluating the effects of steroid hormones on hippocampal neurogenesis.
Collapse
Affiliation(s)
- Travis E Hodges
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
37
|
Salehzadeh M, Soma KK. Glucocorticoid production in the thymus and brain: Immunosteroids and neurosteroids. Brain Behav Immun Health 2021; 18:100352. [PMID: 34988497 PMCID: PMC8710407 DOI: 10.1016/j.bbih.2021.100352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/05/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
Glucocorticoids (GCs) regulate a myriad of physiological systems, such as the immune and nervous systems. Systemic GC levels in blood are often measured as an indicator of local GC levels in target organs. However, several extra-adrenal organs can produce and metabolize GCs locally. More sensitive and specific methods for GC analysis (i.e., mass spectrometry) allow measurement of local GC levels in small tissue samples with low GC concentrations. Consequently, is it now apparent that systemic GC levels often do not reflect local GC levels. Here, we review the use of systemic GC measurements in clinical and research settings, discuss instances where systemic GC levels do not reflect local GC levels, and present evidence that local GC levels provide useful insights, with a focus on local GC production in the thymus (immunosteroids) and brain (neurosteroids). Lastly, we suggest key areas for further research, such as the roles of immunosteroids and neurosteroids in neonatal programming and the potential clinical relevance of local GC modulators.
Collapse
Affiliation(s)
- Melody Salehzadeh
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Kiran K Soma
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
38
|
Chakraborty P, Chattarji S, Jeanneteau F. A salience hypothesis of stress in PTSD. Eur J Neurosci 2021; 54:8029-8051. [PMID: 34766390 DOI: 10.1111/ejn.15526] [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: 07/10/2021] [Revised: 09/13/2021] [Accepted: 10/30/2021] [Indexed: 11/30/2022]
Abstract
Attention to key features of contexts and things is a necessary tool for all organisms. Detecting these salient features of cues, or simply, salience, can also be affected by exposure to traumatic stress, as has been widely reported in individuals suffering from post-traumatic stress disorder (PTSD). Interestingly, similar observations have been robustly replicated across many animal models of stress as well. By using evidence from such rodent stress paradigms, in the present review, we explore PTSD through the lens of salience processing. In this context, we propose that interaction between the neurotrophin brain-derived neurotrophic factor (BDNF) and glucocorticoids determines the long lasting cellular and behavioural consequences of stress salience. We also describe the dual effect of glucocorticoid therapy in the amelioration of PTSD symptoms. Finally, by integrating in vivo observations at multiple scales of plasticity, we propose a unifying hypothesis that pivots on a crucial role of glucocorticoid signalling in dynamically orchestrating stress salience.
Collapse
Affiliation(s)
- Prabahan Chakraborty
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France.,Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India
| | - Sumantra Chattarji
- Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Freddy Jeanneteau
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France
| |
Collapse
|
39
|
Corticosterone in the dorsolateral striatum facilitates the extinction of stimulus-response memory. Neurobiol Learn Mem 2021; 183:107481. [PMID: 34166790 DOI: 10.1016/j.nlm.2021.107481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 11/23/2022]
Abstract
Glucocorticoid hormones are crucially involved in modulating mnemonic processing of stressful or emotionally arousing experiences. They are known to enhance the consolidation of new memories, including those that extinguish older memories. In this study, we investigated whether glucocorticoids facilitate the extinction of a striatum-dependent, and behaviorally more rigid, stimulus-response memory. For this, male rats were initially trained for six days on a stimulus-response task in a T-maze to obtain a reward after making an egocentric right-turn body response, regardless of the starting position in this maze. This training phase was followed by three extinction sessions in which right-turn body responses were not reinforced. Corticosterone administration into the dorsolateral region of the striatum after the first extinction session dose-dependently enhanced the consolidation of extinction memory: Rats administered the higher dose of corticosterone (30 ng), but not lower doses (5 or 10 ng), exhibited significantly fewer right-turn body responses and had longer latencies compared to vehicle-treated animals on the second and third extinction sessions. Co-administration of the glucocorticoid receptor antagonist RU 486 (10 ng) prevented the corticosterone effect, indicating that glucocorticoids enhance the extinction of stimulus-response memory via activation of the glucocorticoid receptor. Corticosterone administration into the dorsomedial striatum did not affect extinction memory. These findings indicate that stress-response mechanisms involving corticosterone actions in the dorsolateral striatum facilitate the extinction of stimulus-response memory that might allow for the development of an opportune behavioral strategy.
Collapse
|
40
|
Tenorio-Lopes L, Kinkead R. Sex-Specific Effects of Stress on Respiratory Control: Plasticity, Adaptation, and Dysfunction. Compr Physiol 2021; 11:2097-2134. [PMID: 34107062 DOI: 10.1002/cphy.c200022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As our understanding of respiratory control evolves, we appreciate how the basic neurobiological principles of plasticity discovered in other systems shape the development and function of the respiratory control system. While breathing is a robust homeostatic function, there is growing evidence that stress disrupts respiratory control in ways that predispose to disease. Neonatal stress (in the form of maternal separation) affects "classical" respiratory control structures such as the peripheral O2 sensors (carotid bodies) and the medulla (e.g., nucleus of the solitary tract). Furthermore, early life stress disrupts the paraventricular nucleus of the hypothalamus (PVH), a structure that has emerged as a primary determinant of the intensity of the ventilatory response to hypoxia. Although underestimated, the PVH's influence on respiratory function is a logical extension of the hypothalamic control of metabolic demand and supply. In this article, we review the functional and anatomical links between the stress neuroendocrine axis and the medullary network regulating breathing. We then present the persistent and sex-specific effects of neonatal stress on respiratory control in adult rats. The similarities between the respiratory phenotype of stressed rats and clinical manifestations of respiratory control disorders such as sleep-disordered breathing and panic attacks are remarkable. These observations are in line with the scientific consensus that the origins of adult disease are often found among developmental and biological disruptions occurring during early life. These observations bring a different perspective on the structural hierarchy of respiratory homeostasis and point to new directions in our understanding of the etiology of respiratory control disorders. © 2021 American Physiological Society. Compr Physiol 11:1-38, 2021.
Collapse
Affiliation(s)
- Luana Tenorio-Lopes
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Richard Kinkead
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
41
|
Renormalizing synapses in sleep: The clock is ticking. Biochem Pharmacol 2021; 191:114533. [PMID: 33771494 DOI: 10.1016/j.bcp.2021.114533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/11/2022]
Abstract
Sleep has been hypothesized to renormalize synapses potentiated in wakefulness. This is proposed to lead to a net reduction in synaptic strength after sleep in brain areas like the cortex and hippocampus. Biological clocks, however, exert independent effects on these synapses that may explain some of the reported differences after wake and sleep. These include changes in synaptic morphology, molecules and efficacy. In this commentary, I discuss why no firm conclusions should be drawn concerning the role of sleep in synaptic renormalization until the role of circadian rhythms are isolated and determined.
Collapse
|
42
|
Tejos-Bravo M, Oakley RH, Whirledge SD, Corrales WA, Silva JP, García-Rojo G, Toledo J, Sanchez W, Román-Albasini L, Aliaga E, Aguayo F, Olave F, Maracaja-Coutinho V, Cidlowski JA, Fiedler JL. Deletion of hippocampal Glucocorticoid receptors unveils sex-biased microRNA expression and neuronal morphology alterations in mice. Neurobiol Stress 2021; 14:100306. [PMID: 33665240 PMCID: PMC7906897 DOI: 10.1016/j.ynstr.2021.100306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 01/18/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Sex differences in the brain have prompted many researchers to investigate the underlying molecular actors, such as the glucocorticoid receptor (GR). This nuclear receptor controls gene expression, including microRNAs (miRNAs), in non-neuronal cells. Here, we investigated sex-biased effects of GR on hippocampal miRNA expression and neuronal morphology by generating a neuron-specific GR knockout mouse (Emx1-Nr3c1−/−). The levels of 578 mature miRNAs were assessed using NanoString technology and, in contrast to males, female Emx1-Nr3c1−/− mice showed a substantially higher number of differentially expressed miRNAs, confirming a sex-biased effect of GR ablation. Based on bioinformatic analyses we identified several transcription factors potentially involved in miRNA regulation. Functional enrichment analyses of the miRNA-mRNA interactions revealed pathways related to neuronal arborization and both spine morphology and density in both sexes. Two recognized regulators of dendritic morphology, CAMKII-α and GSK-3β, increased their protein levels by GR ablation in female mice hippocampus, without changes in males. Additionally, sex-specific effects of GR deletion were observed on CA1 neuronal arborization and dendritic spine features. For instance, a reduced density of mushroom spines in apical dendrites was evidenced only in females, while a decreased length in basal dendrites was noted only in males. However, length and arborization of apical dendrites were reduced by GR ablation irrespective of the sex. Overall, our study provides new insights into the sex-biased GR actions, especially in terms of miRNAs expression and neuronal morphology in the hippocampus.
Collapse
Affiliation(s)
- Macarena Tejos-Bravo
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Robert H Oakley
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, 27709, USA
| | - Shannon D Whirledge
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, 27709, USA
| | - 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
| | - Juan P 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
| | - Gonzalo García-Rojo
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, 8380492, Santiago, Chile.,Carrera de Odontología. Facultad de Ciencias, Universidad de La Serena, La Serena, Chile
| | - Jorge Toledo
- Laboratory of Scientific Image Analysis (SCIAN-Lab), Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Independencia 1027, Santiago, 8380453, Chile
| | - Wendy Sanchez
- Laboratory of Scientific Image Analysis (SCIAN-Lab), Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Independencia 1027, Santiago, 8380453, Chile
| | - Luciano Román-Albasini
- 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
- Department of Kinesiology and the Neuropsychology and Cognitive Neurosciences Research Center (CINPSI-Neurocog), Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile
| | - Felipe Aguayo
- 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 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
| | - Vinicius Maracaja-Coutinho
- 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
| | - John A Cidlowski
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, 27709, USA
| | - 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
|
43
|
Sheng JA, Bales NJ, Myers SA, Bautista AI, Roueinfar M, Hale TM, Handa RJ. The Hypothalamic-Pituitary-Adrenal Axis: Development, Programming Actions of Hormones, and Maternal-Fetal Interactions. Front Behav Neurosci 2021; 14:601939. [PMID: 33519393 PMCID: PMC7838595 DOI: 10.3389/fnbeh.2020.601939] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
The hypothalamic-pituitary-adrenal axis is a complex system of neuroendocrine pathways and feedback loops that function to maintain physiological homeostasis. Abnormal development of the hypothalamic-pituitary-adrenal (HPA) axis can further result in long-term alterations in neuropeptide and neurotransmitter synthesis in the central nervous system, as well as glucocorticoid hormone synthesis in the periphery. Together, these changes can potentially lead to a disruption in neuroendocrine, behavioral, autonomic, and metabolic functions in adulthood. In this review, we will discuss the regulation of the HPA axis and its development. We will also examine the maternal-fetal hypothalamic-pituitary-adrenal axis and disruption of the normal fetal environment which becomes a major risk factor for many neurodevelopmental pathologies in adulthood, such as major depressive disorder, anxiety, schizophrenia, and others.
Collapse
Affiliation(s)
- Julietta A. Sheng
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Natalie J. Bales
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Sage A. Myers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Anna I. Bautista
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Mina Roueinfar
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Taben M. Hale
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Robert J. Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, United States
| |
Collapse
|
44
|
Dudek KA, Dion‐Albert L, Kaufmann FN, Tuck E, Lebel M, Menard C. Neurobiology of resilience in depression: immune and vascular insights from human and animal studies. Eur J Neurosci 2021; 53:183-221. [PMID: 31421056 PMCID: PMC7891571 DOI: 10.1111/ejn.14547] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
Major depressive disorder (MDD) is a chronic and recurrent psychiatric condition characterized by depressed mood, social isolation and anhedonia. It will affect 20% of individuals with considerable economic impacts. Unfortunately, 30-50% of depressed individuals are resistant to current antidepressant treatments. MDD is twice as prevalent in women and associated symptoms are different. Depression's main environmental risk factor is chronic stress, and women report higher levels of stress in daily life. However, not every stressed individual becomes depressed, highlighting the need to identify biological determinants of stress vulnerability but also resilience. Based on a reverse translational approach, rodent models of depression were developed to study the mechanisms underlying susceptibility vs resilience. Indeed, a subpopulation of animals can display coping mechanisms and a set of biological alterations leading to stress resilience. The aetiology of MDD is multifactorial and involves several physiological systems. Exacerbation of endocrine and immune responses from both innate and adaptive systems are observed in depressed individuals and mice exhibiting depression-like behaviours. Increasing attention has been given to neurovascular health since higher prevalence of cardiovascular diseases is found in MDD patients and inflammatory conditions are associated with depression, treatment resistance and relapse. Here, we provide an overview of endocrine, immune and vascular changes associated with stress vulnerability vs. resilience in rodents and when available, in humans. Lack of treatment efficacy suggests that neuron-centric treatments do not address important causal biological factors and better understanding of stress-induced adaptations, including sex differences, could contribute to develop novel therapeutic strategies including personalized medicine approaches.
Collapse
Affiliation(s)
- Katarzyna A. Dudek
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Laurence Dion‐Albert
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Fernanda Neutzling Kaufmann
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Ellen Tuck
- Smurfit Institute of GeneticsTrinity CollegeDublinIreland
| | - Manon Lebel
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Caroline Menard
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| |
Collapse
|
45
|
Effect of childhood physical abuse on social anxiety is mediated via reduced frontal lobe and amygdala-hippocampus complex volume in adult clinical high-risk subjects. Schizophr Res 2021; 227:101-109. [PMID: 32461085 DOI: 10.1016/j.schres.2020.05.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/22/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Childhood adverse experiences (CAE) are associated with clinical psychiatric disorders and symptoms, and with volumetric abnormalities in the amygdala-hippocampus complex (AmHiC) and frontal lobe (FroL) in adulthood. AIM To study whether CAE are associated with reduced AmHiC and FroL and whether these structures mediate the effect of CAE on social anxiety and depression. METHOD In seven European centres, 374 patients with recent onset of psychosis (n = 127), clinical high-risk to psychosis (n = 119) or recent onset of depression (n = 128) were scanned with MRI and their FroL and AmHiC volumes were measured. They all completed self-report scales for assessment of CAE, social anxiety and depression. RESULTS Of the CAE domains, physical abuse was associated specifically with reduced grey and white matter volumes of FroL and AmHiC in psychotic and high-risk patients. After controlling intracranial volume, PhyAb associated significantly with FroL and its grey matter volume in high-risk patients only. In mediation analyses, the effect of physical abuse on social anxiety was mediated via reduced FroL grey mater volume in high-risk patients. In them, when the effects of AmHiC and depression were controlled, the effect of physical abuse on social anxiety was mediated via FroL grey matter volume reduction. CONCLUSIONS Childhood physical abuse is associated with reduced frontal lobe and amygdala-hippocampus complex volume in adult subjects with psychotic symptoms. Reduced frontal lobe and amygdala-hippocampus complex volume mediate the effect of physical abuse on social anxiety in high-risk patients. The effect of physical abuse on depression-independent social anxiety is mediated via reduced frontal lobe.
Collapse
|
46
|
Katan EA. [Neurobiological effects of childhood abuse and neglect as a predisposing factor to substance use disorder]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:141-147. [PMID: 33340309 DOI: 10.17116/jnevro2020120111141] [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: 11/17/2022]
Abstract
The article presents data from research literature on the neurobiological consequences of abuse and neglect in childhood. It is shown that early life stress affects the trajectory of brain development, changes the patterns of cognitive behavioral control and reception of rewards, potentially increasing the risk of substance misuse. The combination of this factor with an immature prefrontal cortex can dramatically increase the liability to the development of substance dependence.
Collapse
Affiliation(s)
- E A Katan
- Orenburg State Medical University, Orenburg, Russia
| |
Collapse
|
47
|
Glushchak K, Ficarro A, Schoenfeld TJ. High-fat diet and acute stress have different effects on object preference tests in rats during adolescence and adulthood. Behav Brain Res 2020; 399:112993. [PMID: 33152318 DOI: 10.1016/j.bbr.2020.112993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/30/2020] [Accepted: 10/28/2020] [Indexed: 12/16/2022]
Abstract
Meals of high-fat diet (HFD) during adolescence produce stronger impairments to memory during adolescence than adulthood, however recovery of memory from adolescent HFD is underexplored. In addition, many tests of rodent memory are confounded by aversive or food-based stimuli, making it difficult to determine baseline memory processing affected by HFD. Thus, we utilized three cohorts of rats (adolescent HFD, adult HFD, and adolescent HFD with recovery) to explore the effects of HFD at different ages on two traditional tests of memory based strictly on object exploration, novel object recognition and novel object location tests. To isolate stress as a variable, rats were tested either at baseline or with cold water swim occurring directly after object acquisition. Results show that preference for novel objects is impaired by stress across all groups, but HFD alone only impairs preference for novel objects during adolescence, although this recovers after switching to a control diet. Additionally, preference for an object in a new location is impaired by HFD in all age groups and fails to recover following diet change. Together the data suggest that stress and HFD differentially affect object preference, based on test type, except during the adolescent period. Because these tests are traditionally interpreted as memory processes dependent on two distinct brain regions, the hippocampus and perirhinal cortex, these results support that stress and HFD affect the hippocampus and perirhinal cortex differently. The data affirm that while perirhinal cortex-dependent behavior recovers, the adolescent period is susceptible to long-lasting dysfunctions of hippocampal behavior by HFD.
Collapse
Affiliation(s)
- Karina Glushchak
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, TN, 37212, USA
| | - Alexandria Ficarro
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, TN, 37212, USA
| | - Timothy J Schoenfeld
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, TN, 37212, USA.
| |
Collapse
|
48
|
Razavi Y, Karimi S, Karimi-Haghighi S, Hesam S, Haghparast A. Changes in c-fos and p-CREB signaling following exposure to forced swim stress or exogenous corticosterone during morphine-induced place preference are dependent on glucocorticoid receptor in the basolateral amygdala. Can J Physiol Pharmacol 2020; 98:741-752. [PMID: 32574519 DOI: 10.1139/cjpp-2019-0712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neural circuitry comprising the nucleus accumbens (NAc), prefrontal cortex (PFC), amygdala (AMY), and hippocampus (HIP) are the main components of the reward circuit. Our previous behavioral data showed that forced swim stress (FSS) and corticosterone administration could inhibit the acquisition of morphine-induced conditioned place preference (CPP), and this effect was blocked by intra-basolateral amygdala (BLA) administration of RU38486, glucocorticoid receptor (GR) antagonist. Therefore, we tried to evaluate the effect of intra-BLA administration of the GR antagonist during the conditioning phase on the c-fos and p-CREB/CREB ratio expression in the AMY, NAc, PFC, and HIP of rats that underwent FSS or received exogenous corticosterone (10 mg/kg; i.p.) before morphine injection (5 mg/kg; s.c.) during 3 conditioning days. Our results showed that morphine-induced CPP could increase c-fos level and p-CREB/CREB ratio in all regions (except in the HIP). In addition, c-fos expression was elevated by FSS in all regions and blockade of GR decreased this effect. In the PFC, in addition to FSS, corticosterone could raise c-fos expression, which was blocked by RU38486. In conclusion, it seems that the intra-BLA administration of RU38486 differently modulates the effect of morphine-induced CPP on the expression of c-fos and p-CREB/CREB ratio in animals that underwent FSS or corticosterone administration.
Collapse
Affiliation(s)
- Yasaman Razavi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Karimi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeideh Karimi-Haghighi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soghra Hesam
- Department of Neuroscience, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Boorman DC, Brown R, Keay KA. Periaqueductal gray inputs to the paraventricular nucleus of the thalamus: Columnar topography and glucocorticoid (in)sensitivity. Brain Res 2020; 1750:147171. [PMID: 33132167 DOI: 10.1016/j.brainres.2020.147171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/10/2020] [Accepted: 10/18/2020] [Indexed: 01/01/2023]
Abstract
The ability to cope with a novel acute stressor in the context of ongoing chronic stress is of critical adaptive value. The hypothalamic-pituitary-adrenal (HPA) axis contributes to the integrated physiological and behavioural responses to stressors. Under conditions of chronic stress, the posterior portion of the paraventricular thalamic nucleus (pPVT) mediates the 'habituation' of HPA-axis responses, and also facilitates HPA-axis reactivation to novel acute stressors amidst this habituation. Since pPVT neurons are sensitive to the inhibitory effects of circulating glucocorticoids, a glucocorticoid-insensitive neural pathway to the pPVT is likely essential for this reactivation process. The pPVT receives substantial inputs from neurons of the periaqueductal gray (PAG) region, which is organised into longitudinal columns critical for processing acute and/or chronic stressors. We investigated the columnar organisation of PAG → pPVT projections and for the first time determined their glucocorticoid sensitivity. Retrograde tracer injections were made into different rostro-caudal regions of the pPVT, and their PAG columnar inputs compared. Glucocorticoid receptor immunoreactivity (GR-ir) was quantified in these projection neurons. We found that the dorsolateral PAG projected most strongly to rostral pPVT and the ventrolateral PAG most strongly to the caudal pPVT. Despite abundant GR-ir in the PAG, we report a striking absence of GR-ir in PAG → pPVT neurons. Our data suggests that these pathways, which are insensitive to the direct actions of circulating glucocorticoids, likely play an important role in both the habituation of HPA-axis to chronic stressors and its facilitation to acute stressors in chronically stressed rats.
Collapse
Affiliation(s)
- Damien C Boorman
- School of Medical Sciences (Anatomy & Histology), Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Rebecca Brown
- School of Medical Sciences (Anatomy & Histology), Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Kevin A Keay
- School of Medical Sciences (Anatomy & Histology), Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
50
|
Ekizceli G, Halk KZ, Minbay Z, Eyigor O. Nesfatin-1 and neuronostatin neurons are co-expressed with glucocorticoid receptors in the hypothalamus. Biotech Histochem 2020; 96:555-561. [PMID: 33054452 DOI: 10.1080/10520295.2020.1832703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Nesfatin-1 and neuronostatin in the central nervous system participate in regulating stress responses. Glucocorticoid hormones affect the brain through glucocorticoid receptors (GR). We investigated in the rat the possibility of co-localizing nesfatin-1 and neuronostatin neurons in hypothalamic areas with GR. using immunohistochemistry. We counted nesfatin-1 and neuronostatin stained neurons. We counted GR positive nesfatin-1 neurons in the arcuate nucleus (ARC) and paraventricular nucleus (PVN) and GR positive neuronostatin neurons in the periventricular nucleus (PeN). The percentage of nesfatin-1 neurons that expressed GR was 38.4% in female rats and 21.9% in male rats in the ARC, and 33.3% in female rats and 29.2% in male rats in the PVN. The percentage of neuronostatin neurons that expressed GR was 39.1% in female rats and 39.9% in male rats in the PeN. We found that a substantial portion of nesfatin-1 and neuronostatin neurons were stained for GR. We speculate that the pattern of GR might permit secretion of neuropeptides to be stimulated by peripheral glucocorticoid signals. Stress can suppress food intake, in part, through the GR in neurons that express nesfatin-1, which is a satiety molecule, and in neurons that express neuronostatin, which is an anorexigenic peptide.
Collapse
Affiliation(s)
- G Ekizceli
- Department of Histology and Embryology, Istanbul Health and Technology University, School of Medicine, Istanbul, Turkey.,Department of Histology and Embryology, Bursa Uludag University, Institute of Health Science, Bursa, Turkey
| | - K Z Halk
- Department of Histology and Embryology, Bursa Uludag University, Institute of Health Science, Bursa, Turkey
| | - Z Minbay
- Department of Histology and Embryology, Bursa Uludag University, School of Medicine, Bursa, Turkey
| | - O Eyigor
- Department of Histology and Embryology, Bursa Uludag University, School of Medicine, Bursa, Turkey
| |
Collapse
|