1
|
Zuo Z, Zhang H, Li Z, Qi F, Hu H, Yang J, Yao Z. Activation of Hippocampal Neuronal NADPH Oxidase NOX2 Promotes Depressive-Like Behaviour and Cognition Deficits in Chronic Restraint Stress Mouse Model. PHARMACOPSYCHIATRY 2025; 58:117-126. [PMID: 39547705 DOI: 10.1055/a-2429-4023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
BACKGROUND Nicotinamide adenosine dinucleotide phosphate oxidases (NOX) play important roles in mediating stress-induced depression. Three NOX isotypes are expressed mainly in the brain: NOX2, NOX3 and NOX4. In this study, the expression and cellular sources of these NOX isoforms was investigated in the context of stress-induced depression. METHODS Chronic restraint stress (CRS)-induced depressive-like behaviour and cognitive deficits were evaluated by tail suspension tests, forced swimming tests and the Morris water maze test. Hippocampal NOX expression was determined by immunofluorescence staining and western blotting. The hippocampal levels of the brain-derived neurotrophic factor (BDNF) mRNA were determined via quantitative real-time -polymerase chain reaction. Glucocorticoid levels in the hippocampus were measured using ELISA kits. RESULTS In the mouse CRS model, a significant increase in NOX2 expression was observed in the hippocampus, whereas no significant changes in NOX3 and NOX4 expression were detected. Next, NOX2 expression was primarily localised to neurons (NeuN+) but not microglia (Iba-1+) or astrocytes (GFAP+). Treatment with gp91ds-tat, a specific NOX2 inhibitor, effectively mitigated the behavioural deficits induced by CRS. The decreased expression of the BDNF mRNA in the hippocampus of CRS mice was restored upon gp91ds-tat treatment. A positive correlation was identified between neuronal NOX2 expression and serum glucocorticoid levels. CONCLUSIONS Our study indicated that neuronal NOX2 may be a critical mediator of depression-like behaviours and spatial cognitive deficits in mice subjected to CRS. Blockade of NOX2 signalling may be a promising therapeutic strategy for depression.
Collapse
Affiliation(s)
- Zejie Zuo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongyang Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhihui Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fangfang Qi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Haojie Hu
- Department of Psychology, College of Arts and Sciences, New York University, NY, USA
| | - Junhua Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhibin Yao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Martínez-Martos JM, Cantón-Habas V, Rich-Ruíz M, Reyes-Medina MJ, Ramírez-Expósito MJ, Carrera-González MDP. Sexual and Metabolic Differences in Hippocampal Evolution: Alzheimer's Disease Implications. Life (Basel) 2024; 14:1547. [PMID: 39768255 PMCID: PMC11677427 DOI: 10.3390/life14121547] [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: 10/25/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Sex differences in brain metabolism and their relationship to neurodegenerative diseases like Alzheimer's are an important emerging topic in neuroscience. Intrinsic anatomic and metabolic differences related to male and female physiology have been described, underscoring the importance of considering biological sex in studying brain metabolism and associated pathologies. The hippocampus is a key structure exhibiting sex differences in volume and connectivity. Adult neurogenesis in the dentate gyrus, dendritic spine density, and electrophysiological plasticity contribute to the hippocampus' remarkable plasticity. Glucose transporters GLUT3 and GLUT4 are expressed in human hippocampal neurons, with proper glucose metabolism being crucial for learning and memory. Sex hormones play a major role, with the aromatase enzyme that generates estradiol increasing in neurons and astrocytes as an endogenous neuroprotective mechanism. Inhibition of aromatase increases gliosis and neurodegeneration after brain injury. Genetic variants of aromatase may confer higher Alzheimer's risk. Estrogen replacement therapy in postmenopausal women prevents hippocampal hypometabolism and preserves memory. Insulin is also a key regulator of hippocampal glucose metabolism and cognitive processes. Dysregulation of the insulin-sensitive glucose transporter GLUT4 may explain the comorbidity between type II diabetes and Alzheimer's. GLUT4 colocalizes with the insulin-regulated aminopeptidase IRAP in neuronal vesicles, suggesting an activity-dependent glucose uptake mechanism. Sex differences in brain metabolism are an important factor in understanding neurodegenerative diseases, and future research must elucidate the underlying mechanisms and potential therapeutic implications of these differences.
Collapse
Affiliation(s)
- José Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Las Lagunillas University Campus, 23009 Jaen, Spain; (J.M.M.-M.); (M.J.R.-E.)
| | - Vanesa Cantón-Habas
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, University of Córdoba, 14004 Córdoba, Spain; (V.C.-H.); (M.R.-R.); (M.J.R.-M.)
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14004 Cordoba, Spain
| | - Manuel Rich-Ruíz
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, University of Córdoba, 14004 Córdoba, Spain; (V.C.-H.); (M.R.-R.); (M.J.R.-M.)
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14004 Cordoba, Spain
| | - María José Reyes-Medina
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, University of Córdoba, 14004 Córdoba, Spain; (V.C.-H.); (M.R.-R.); (M.J.R.-M.)
| | - María Jesús Ramírez-Expósito
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Las Lagunillas University Campus, 23009 Jaen, Spain; (J.M.M.-M.); (M.J.R.-E.)
| | - María del Pilar Carrera-González
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Las Lagunillas University Campus, 23009 Jaen, Spain; (J.M.M.-M.); (M.J.R.-E.)
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14004 Cordoba, Spain
| |
Collapse
|
3
|
Burke MR, Sotiropoulos I, Waites CL. The multiple roles of chronic stress and glucocorticoids in Alzheimer's disease pathogenesis. Trends Neurosci 2024; 47:933-948. [PMID: 39307629 PMCID: PMC11563862 DOI: 10.1016/j.tins.2024.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 11/15/2024]
Abstract
Chronic stress and the accompanying long-term elevation of glucocorticoids (GCs), the stress hormones of the body, increase the risk and accelerate the progression of Alzheimer's disease (AD). Signatures of AD include intracellular tau (MAPT) tangles, extracellular amyloid β (Aβ) plaques, and neuroinflammation. A growing body of work indicates that stress and GCs initiate cellular processes underlying these pathologies through dysregulation of protein homeostasis and trafficking, mitochondrial bioenergetics, and response to damage-associated stimuli. In this review, we integrate findings from mechanistic studies in rodent and cellular models, wherein defined chronic stress protocols or GC administration have been shown to elicit AD-related pathology. We specifically discuss the effects of chronic stress and GCs on tau pathogenesis, including hyperphosphorylation, aggregation, and spreading, amyloid precursor protein (APP) processing and trafficking culminating in Aβ production, immune priming by proinflammatory cytokines and disease-associated molecular patterns, and alterations to glial cell and blood-brain barrier (BBB) function.
Collapse
Affiliation(s)
- Mia R Burke
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Pathobiology and Mechanisms of Disease Graduate Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Ioannis Sotiropoulos
- Institute of Biosciences and Applications, National Centre for Scientific Research (NCSR) Demokritos, Agia Paraskevi, Greece
| | - Clarissa L Waites
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Department of Neuroscience, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Nakos Bimpos M, Karali K, Antoniou C, Palermos D, Fouka M, Delis A, Tzieras I, Chrousos GP, Koutmani Y, Stefanis L, Polissidis A. Alpha-synuclein-induced stress sensitivity renders the Parkinson's disease brain susceptible to neurodegeneration. Acta Neuropathol Commun 2024; 12:100. [PMID: 38886854 PMCID: PMC11181569 DOI: 10.1186/s40478-024-01797-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/12/2024] [Indexed: 06/20/2024] Open
Abstract
A link between chronic stress and Parkinson's disease (PD) pathogenesis is emerging. Ample evidence demonstrates that the presynaptic neuronal protein alpha-synuclein (asyn) is closely tied to PD pathogenesis. However, it is not known whether stress system dysfunction is present in PD, if asyn is involved, and if, together, they contribute to neurodegeneration. To address these questions, we assess stress axis function in transgenic rats overexpressing full-length wildtype human asyn (asyn BAC rats) and perform multi-level stress and PD phenotyping following chronic corticosterone administration. Stress signaling, namely corticotropin-releasing factor, glucocorticoid and mineralocorticoid receptor gene expression, is also examined in post-mortem PD patient brains. Overexpression of human wildtype asyn leads to HPA axis dysregulation in rats, while chronic corticosterone administration significantly aggravates nigrostriatal degeneration, serine129 phosphorylated asyn (pS129) expression and neuroinflammation, leading to phenoconversion from a prodromal to an overt motor PD phenotype. Interestingly, chronic corticosterone in asyn BAC rats induces a robust, twofold increase in pS129 expression in the hypothalamus, the master regulator of the stress response, while the hippocampus, both a regulator and a target of the stress response, also demonstrates elevated pS129 asyn levels and altered markers of stress signalling. Finally, defective hippocampal stress signalling is mirrored in human PD brains and correlates with asyn expression levels. Taken together, our results link brain stress system dysregulation with asyn and provide evidence that elevated circulating glucocorticoids can contribute to asyn-induced neurodegeneration, ultimately triggering phenoconversion from prodromal to overt PD.
Collapse
Affiliation(s)
- Modestos Nakos Bimpos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Katerina Karali
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- German Center for Neurodegenerative Diseases, Feodor-Lynen-Straße 17, 81377, Munich, Germany
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, 15784, Illisia, Athens, Greece
| | - Christine Antoniou
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, 15784, Illisia, Athens, Greece
| | - Dionysios Palermos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Maria Fouka
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Anastasios Delis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Iason Tzieras
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - George Panagiotis Chrousos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- University Research Institute on Maternal and Child Health and Precision Medicine, and UNESCO Chair on Adolescent Health Care, Aghia Sophia Children's Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Yassemi Koutmani
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
| | - Leonidas Stefanis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece
- 1St Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Alexia Polissidis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens - BRFAA, 11527, Athens, Greece.
- Department of Science and Mathematics, ACG-Research Center, Deree - American College of Greece, 15342, Athens, Greece.
| |
Collapse
|
7
|
Kim S, Yang S, Kim J, Chung KW, Jung YS, Chung HY, Lee J. Glucocorticoid Receptor Down-Regulation Affects Neural Stem Cell Proliferation and Hippocampal Neurogenesis. Mol Neurobiol 2024; 61:3198-3211. [PMID: 37979034 DOI: 10.1007/s12035-023-03785-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Dysregulation of the hypothalamic-pituitary-adrenal axis and abnormalities in the glucocorticoid receptor (GR) have been linked to major depressive disorder. Given the critical role of GR in stress response regulation, we investigated the impact of GR changes on neural stem cells (NSCs) proliferation and hippocampal neurogenesis. Stress response was induced using dexamethasone (DEX), a GR agonist, which led to reduced proliferation of neural stem cells and neural progenitor cells, as well as decreased expression of GR. Additionally, a reduction of serum concentration within the culture media resulted in suppressed cell proliferation, accompanied by decreased GR expression. The association between GR expression and cell proliferation was further confirmed through GR siRNA knockdown and overexpression experiments. Furthermore, in vivo studies utilizing young male C57BL/6 mice demonstrated that corticosterone (CORT) (35 μg/ml) administered through drinking water for four weeks induced depression-like behavior, as indicated by increased immobility times in forced swimming and tail suspension tests. CORT exposure led to reduced GR and nestin expression levels, along with diminished numbers of BrdU-positive cells in the hippocampi, indicating impaired hippocampal neurogenesis. Taken together, our findings provide the first evidence that stress-induced downregulation of GR negatively affects neurogenesis by inhibiting NSCs proliferation.
Collapse
Affiliation(s)
- Seoyeong Kim
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, 46241, Republic of Korea
| | - Seonguk Yang
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, 46241, Republic of Korea
| | - Jaehoon Kim
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, 46241, Republic of Korea
| | - Ki Wung Chung
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, 46241, Republic of Korea
| | - Young-Suk Jung
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, 46241, Republic of Korea
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, 46241, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
8
|
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
|
9
|
Chamaa F, Magistretti PJ, Fiumelli H. Astrocyte-derived lactate in stress disorders. Neurobiol Dis 2024; 192:106417. [PMID: 38296112 DOI: 10.1016/j.nbd.2024.106417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/04/2023] [Accepted: 01/23/2024] [Indexed: 02/05/2024] Open
Abstract
Stress disorders are psychiatric disorders arising following stressful or traumatic events. They could deleteriously affect an individual's health because they often co-occur with mental illnesses. Considerable attention has been focused on neurons when considering the neurobiology of stress disorders. However, like other mental health conditions, recent studies have highlighted the importance of astrocytes in the pathophysiology of stress-related disorders. In addition to their structural and homeostatic support role, astrocytes actively serve several functions in regulating synaptic transmission and plasticity, protecting neurons from toxic compounds, and providing metabolic support for neurons. The astrocyte-neuron lactate shuttle model sets forth the importance of astrocytes in providing lactate for the metabolic supply of neurons under intense activity. Lactate also plays a role as a signaling molecule and has been recently studied regarding its antidepressant activity. This review discusses the involvement of astrocytes and brain energy metabolism in stress and further reflects on the importance of lactate as an energy supply in the brain and its emerging antidepressant role in stress-related disorders.
Collapse
Affiliation(s)
- Farah Chamaa
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Pierre J Magistretti
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Hubert Fiumelli
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia.
| |
Collapse
|
10
|
Wei B, Shi H, Yu X, Shi Y, Zeng H, Zhao Y, Zhao Z, Song Y, Sun M, Wang B. GR/Ahi1 regulates WDR68-DYRK1A binding and mediates cognitive impairment in prenatally stressed offspring. Cell Mol Life Sci 2024; 81:20. [PMID: 38195774 PMCID: PMC11073104 DOI: 10.1007/s00018-023-05075-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/19/2023] [Accepted: 11/29/2023] [Indexed: 01/11/2024]
Abstract
Accumulating research shows that prenatal exposure to maternal stress increases the risk of behavioral and mental health problems for offspring later in life. However, how prenatal stress affects offspring behavior remains unknown. Here, we found that prenatal stress (PNS) leads to reduced Ahi1, decreased synaptic plasticity and cognitive impairment in offspring. Mechanistically, Ahi1 and GR stabilize each other, inhibit GR nuclear translocation, promote Ahi1 and WDR68 binding, and inhibit DYRK1A and WDR68 binding. When Ahi1 deletion or prenatal stress leads to hyperactivity of the HPA axis, it promotes the release of GC, leading to GR nuclear translocation and Ahi1 degradation, which further inhibits the binding of Ahi1 and WDR68, and promotes the binding of DYRK1A and WDR68, leading to elevated DYRK1A, reduced synaptic plasticity, and cognitive impairment. Interestingly, we identified RU486, an antagonist of GR, which increased Ahi1/GR levels and improved cognitive impairment and synaptic plasticity in PNS offspring. Our study contributes to understanding the signaling mechanisms of prenatal stress-mediated cognitive impairment in offspring.
Collapse
Affiliation(s)
- Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Haixia Shi
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xi Yu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Hongtao Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zejun Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yueyang Song
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Bin Wang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
11
|
Phillips NS, Mulrooney DA, Williams AM, Liu W, Khan RB, Ehrhardt MJ, Folse T, Krasin M, Srivastava DK, Ness KK, Hudson MM, Sabin ND, Krull KR. Neurocognitive impairment associated with chronic morbidity in long-term survivors of Hodgkin Lymphoma. Blood Adv 2023; 7:7270-7278. [PMID: 37729618 PMCID: PMC10711168 DOI: 10.1182/bloodadvances.2023010567] [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: 04/26/2023] [Revised: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
Thoracic radiation is associated with significant cardiopulmonary morbidities in survivors of long-term Hodgkin lymphoma and may affect neurocognitive outcomes. Survivors (N = 204; 52.5% female; mean [standard deviation] age, 36.6 [8.01] years) treated with thoracic radiation and age-, sex-, and race/ethnicity-matched community controls (N = 205; 51.7% female; age, 36.7 [9.17] years) completed standardized neurocognitive testing, echocardiography, pulmonary function tests, and vascular studies during the same visit. Treatments were abstracted from medical records. Cardiac (ie, left ventricular ejection fraction [LVEF], global longitudinal strain [GLS]), vascular (ie, large and small artery elasticity [SAE]), pulmonary (ie, diffusing capacity of the lungs for carbon monoxide [DLCO] and forced expiratory volume [FEV1]), and chronic health conditions were evaluated for associations with age-adjusted neurocognitive performance using multivariable linear regression. Compared with controls, survivors had lower performance (P < 0.05) in visuomotor (0.11 vs 0.41), visual processing speed (0.25 vs 0.64), short-term recall (-0.24 vs 0.12), and flexibility (-0.04 vs 0.28). Survivors had lower pulmonary (FEV1, DLCOcorr), cardiac (LVEF, GLS), and vascular function (SAE) than controls (all P < 0.001). FEV1 was associated with visuomotor (P = .008) and visual processing speed (P = .05), and flexibility (P = .05). GLS was associated with short-term recall (P = .03). SAE was associated with flexibility (P = .007). Neurocognitive outcomes were also associated with moderate-to-severe neurologic chronic conditions (P < .05). Findings suggest a link between subclinical cardiopulmonary and vascular findings, neurologic morbidity, and neurocognitive impairments. Prevention of health morbidity may benefit neurocognitive outcomes.
Collapse
Affiliation(s)
- Nicholas S. Phillips
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Daniel A. Mulrooney
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - AnnaLynn M. Williams
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Wei Liu
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Raja B. Khan
- Department of Pediatric Medicine Division of Neurology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Matthew J. Ehrhardt
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Tim Folse
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Matthew Krasin
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | | | - Kirsten K. Ness
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Melissa M. Hudson
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Noah D. Sabin
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kevin R. Krull
- Department of Psychology and Biobehavioral Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
12
|
Sherman BE, Harris BB, Turk-Browne NB, Sinha R, Goldfarb EV. Hippocampal Mechanisms Support Cortisol-Induced Memory Enhancements. J Neurosci 2023; 43:7198-7212. [PMID: 37813570 PMCID: PMC10601369 DOI: 10.1523/jneurosci.0916-23.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/05/2023] [Accepted: 09/09/2023] [Indexed: 10/17/2023] Open
Abstract
Stress can powerfully influence episodic memory, often enhancing memory encoding for emotionally salient information. These stress-induced memory enhancements stand at odds with demonstrations that stress and the stress-related hormone cortisol can negatively affect the hippocampus, a brain region important for episodic memory encoding. To resolve this apparent conflict and determine whether and how the hippocampus supports memory encoding under cortisol, we combined behavioral assays of associative memory, high-resolution fMRI, and pharmacological manipulation of cortisol in a within-participant, double-blinded procedure (in both sexes). Behaviorally, hydrocortisone promoted the encoding of subjectively arousing, positive associative memories. Neurally, hydrocortisone led to enhanced functional connectivity between hippocampal subregions, which predicted subsequent memory enhancements for emotional associations. Cortisol also modified the relationship between hippocampal representations and associative memory: whereas hippocampal signatures of distinctiveness predicted memory under placebo, relative integration predicted memory under cortisol. Together, these data provide novel evidence that the human hippocampus contains the necessary machinery to support emotional associative memory enhancements under cortisol.SIGNIFICANCE STATEMENT Our daily lives are filled with stressful events, which powerfully shape the way we form episodic memories. For example, stress and stress-related hormones can enhance our memory for emotional events. However, the mechanisms underlying these memory benefits are unclear. In the current study, we combined functional neuroimaging, behavioral tests of memory, and double-blind, placebo-controlled hydrocortisone administration to uncover the effects of the stress-related hormone cortisol on the function of the human hippocampus, a brain region important for episodic memory. We identified novel ways in which cortisol can enhance hippocampal function to promote emotional memories, highlighting the adaptive role of cortisol in shaping memory formation.
Collapse
Affiliation(s)
- Brynn E Sherman
- Department of Psychology, University of Pennsylvania, Philadelphia 19104
| | - Bailey B Harris
- Department of Psychology, UCLA, Los Angeles, California 90095
| | - Nicholas B Turk-Browne
- Department of Psychology, Yale University, New Haven, Connecticut 06520
- Wu Tsai Institute, Yale University, New Haven, Connecticut 06510
| | - Rajita Sinha
- Department of Psychiatry, Yale University, New Haven, Connecticut 06511
| | - Elizabeth V Goldfarb
- Department of Psychology, Yale University, New Haven, Connecticut 06520
- Wu Tsai Institute, Yale University, New Haven, Connecticut 06510
- Department of Psychiatry, Yale University, New Haven, Connecticut 06511
- National Center for PTSD, VA Connecticut Healthcare System, West Haven, Connecticut 06477
| |
Collapse
|
13
|
Jue H, Fang-fang L, Dan-fei C, Nuo C, Chun-lu Y, Ke-pin Y, Jian C, Xiao-bo X. A bidirectional Mendelian randomization study about the role of morning plasma cortisol in attention deficit hyperactivity disorder. Front Psychiatry 2023; 14:1148759. [PMID: 37389173 PMCID: PMC10303788 DOI: 10.3389/fpsyt.2023.1148759] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/29/2023] [Indexed: 07/01/2023] Open
Abstract
Context Cortisol, a hormone regulated by the hypothalamic-pituitary-adrenal (HPA) axis, has been linked to attention deficit hyperactivity disorder (ADHD). The nature of the relationship between cortisol and ADHD, and whether it is causal or explained by reverse causality, remains a matter of debate. Objective This study aims to evaluate the bidirectional causal relationship between morning plasma cortisol levels and ADHD. Methods This study used a bidirectional 2-sample Mendelian randomization (MR) design to analyze the association between morning plasma cortisol levels and ADHD using genetic information from the authoritative Psychiatric Genomics Collaboration (PGC) database (n = 55,347) and the ADHD Working Group of the CORtisol NETwork (CORNET) Consortium (n = 12,597). MR analyses were employed: inverse variance weighting (IVW), MR-Egger regression, and weighted medians. OR values and 95% CI were used to evaluate whether there was a causal association between morning plasma cortisol levels on ADHD and ADHD on morning plasma cortisol levels. The Egger-intercept method was employed to test for level pleiotropy. Sensitivity analysis was performed using the "leave-one-out" method, MR pleiotropy residual sum, and MR pleiotropy residual sum and outlier (MR-PRESSO). Results Findings from bidirectional MR demonstrated that lower morning plasma cortisol levels were associated with ADHD (ADHD-cortisol OR = 0.857; 95% CI, 0.755-0.974; P = 0.018), suggesting there is a reverse causal relationship between cortisol and ADHD. However, morning plasma cortisol levels were not found to have a causal effect on the risk of ADHD (OR = 1.006; 95% CI, 0.909-1.113; P = 0.907), despite the lack of genetic evidence. The MR-Egger method revealed intercepts close to zero, indicating that the selected instrumental variables had no horizontal multiplicity. The "leave-one-out" sensitivity analysis revealed stable results, with no instrumental variables significantly affecting the results. Heterogeneity tests were insignificant, and MR-PRESSO did not detect any significant outliers. The selected single-nucleotide polymorphisms (SNPs) F were all >10, indicating no weak instrumental variables. Thus, the overall MR analysis results were reliable. Conclusion The study findings suggest a reverse causal relationship between morning plasma cortisol levels and ADHD, with low cortisol levels associated with ADHD. No genetic evidence was found to support a causal relationship between morning plasma cortisol levels and the risk of ADHD. These results suggest that ADHD may lead to a significant reduction in morning plasma cortisol secretion.
Collapse
Affiliation(s)
- Hu Jue
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Fang-fang
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chen Dan-fei
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chen Nuo
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ye Chun-lu
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Ke-pin
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chen Jian
- First Clinical School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuan Xiao-bo
- Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
14
|
Afridi R, Suk K. Microglial Responses to Stress-Induced Depression: Causes and Consequences. Cells 2023; 12:1521. [PMID: 37296642 PMCID: PMC10252665 DOI: 10.3390/cells12111521] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Chronic stress is a major risk factor for various psychiatric diseases, including depression; it triggers various cellular and structural changes, resulting in the alteration of neurocircuitry and subsequent development of depression. Accumulating evidence suggests that microglial cells orchestrate stress-induced depression. Preclinical studies of stress-induced depression revealed microglial inflammatory activation in regions of the brain that regulate mood. Although studies have identified several molecules that trigger inflammatory responses in microglia, the pathways that regulate stress-induced microglial activation remain unclear. Understanding the exact triggers that induce microglial inflammatory activation can help find therapeutic targets in order to treat depression. In the current review, we summarize the recent literature on possible sources of microglial inflammatory activation in animal models of chronic stress-induced depression. In addition, we describe how microglial inflammatory signaling affects neuronal health and causes depressive-like behavior in animal models. Finally, we propose ways to target the microglial inflammatory cascade to treat depressive disorders.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu 41940, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu 41940, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
15
|
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
|
16
|
Tran KN, Nguyen NPK, Nguyen LTH, Shin HM, Yang IJ. Screening for Neuroprotective and Rapid Antidepressant-like Effects of 20 Essential Oils. Biomedicines 2023; 11:biomedicines11051248. [PMID: 37238920 DOI: 10.3390/biomedicines11051248] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Depression is a serious psychiatric disorder with high prevalence, and the delayed onset of antidepressant effects remains a limitation in the treatment of depression. This study aimed to screen essential oils that have the potential for rapid-acting antidepressant development. PC12 and BV2 cells were used to identify essential oils with neuroprotective effects at doses of 0.1 and 1 µg/mL. The resulting candidates were treated intranasally (25 mg/kg) to ICR mice, followed by a tail suspension test (TST) and an elevated plus maze (EPM) after 30 min. In each effective essential oil, five main compounds were computationally analyzed, targeting glutamate receptor subunits. As a result, 19 essential oils significantly abolished corticosterone (CORT)-induced cell death and lactate dehydrogenase (LDH) leakage, and 13 reduced lipopolysaccharide (LPS)-induced tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6). From in vivo experiments, six essential oils decreased the immobility time of mice in the TST, in which Chrysanthemum morifolium Ramat. and Myristica fragrans Houtt. also increased time and entries into the open arms of the EPM. Four compounds including atractylon, α-curcumene, α-farnesene, and selina-4(14),7(11)-dien-8-one had an affinity toward GluN1, GluN2B, and Glu2A receptor subunits surpassed that of the reference compound ketamine. Overall, Atractylodes lancea (Thunb.) DC and Chrysanthemum morifolium Ramat essential oils are worthy of further research for fast-acting antidepressants through interactions with glutamate receptors, and their main compounds (atractylon, α-curcumene, α-farnesene, and selina-4(14),7(11)-dien-8-one) are predicted to underlie the fast-acting effect.
Collapse
Affiliation(s)
- Khoa Nguyen Tran
- Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Nhi Phuc Khanh Nguyen
- Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Ly Thi Huong Nguyen
- Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Heung-Mook Shin
- Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - In-Jun Yang
- Department of Physiology, College of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| |
Collapse
|
17
|
Teo CH, Wong ACH, Sivakumaran RN, Parhar I, Soga T. Gender Differences in Cortisol and Cortisol Receptors in Depression: A Narrative Review. Int J Mol Sci 2023; 24:ijms24087129. [PMID: 37108291 PMCID: PMC10138698 DOI: 10.3390/ijms24087129] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Stress is known to have a significant impact on mental health. While gender differences can be found in stress response and mental disorders, there are limited studies on the neuronal mechanisms of gender differences in mental health. Here, we discuss gender and cortisol in depression as presented by recent clinical studies, as well as gender differences in the role of glucocorticoid receptors (GRs) and mineralocorticoid receptors (MRs) in stress-associated mental disorders. When examining clinical studies drawn from PubMed/MEDLINE (National Library of Medicine) and EMBASE, salivary cortisol generally showed no gender correlation. However, young males were reported to show heightened cortisol reactivity compared to females of similar age in depression. Pubertal hormones, age, early life stressors, and types of bio-samples for cortisol measurement affected the recorded cortisol levels. The role of GRs and MRs in the HPA axis could be different between males and females during depression, with increased HPA activity and upregulated MR expression in male mice, while the inverse happened in female mice. The functional heterogeneity and imbalance of GRs and MRs in the brain may explain gender differences in mental disorders. This knowledge and understanding will support the development of gender-specific diagnostic markers involving GRs and MRs in depression.
Collapse
Affiliation(s)
- Chuin Hau Teo
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| | - Ally Chai Hui Wong
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| | - Rooba Nair Sivakumaran
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| | - Ishwar Parhar
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| | - Tomoko Soga
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| |
Collapse
|
18
|
Lisakovska O, Labudzynskyi D, Khomenko A, Isaev D, Savotchenko A, Kasatkina L, Savosko S, Veliky M, Shymanskyi I. Brain vitamin D3-auto/paracrine system in relation to structural, neurophysiological, and behavioral disturbances associated with glucocorticoid-induced neurotoxicity. Front Cell Neurosci 2023; 17:1133400. [PMID: 37020845 PMCID: PMC10067932 DOI: 10.3389/fncel.2023.1133400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/28/2023] [Indexed: 03/22/2023] Open
Abstract
IntroductionVitamin D3 (VD3) is a potent para/autocrine regulator and neurosteroid that can strongly influence nerve cell function and counteract the negative effects of glucocorticoid (GC) therapy. The aim of the study was to reveal the relationship between VD3 status and behavioral, structural-functional and molecular changes associated with GC-induced neurotoxicity.MethodsFemale Wistar rats received synthetic GC prednisolone (5 mg/kg b.w.) with or without VD3 (1000 IU/kg b.w.) for 30 days. Behavioral, histological, physiological, biochemical, molecular biological (RT-PCR, Western blotting) methods, and ELISA were used.Results and discussionThere was no difference in open field test (OFT), while forced swim test (FST) showed an increase in immobility time and a decrease in active behavior in prednisolone-treated rats, indicative of depressive changes. GC increased the perikaryon area, enlarged the size of the nuclei, and caused a slight reduction of cell density in CA1-CA3 hippocampal sections. We established a GC-induced decrease in the long-term potentiation (LTP) in CA1-CA3 hippocampal synapses, the amplitude of high K+-stimulated exocytosis, and the rate of Ca2+-dependent fusion of synaptic vesicles with synaptic plasma membranes. These changes were accompanied by an increase in nitration and poly(ADP)-ribosylation of cerebral proteins, suggesting the development of oxidative-nitrosative stress. Prednisolone upregulated the expression and phosphorylation of NF-κB p65 subunit at Ser311, whereas downregulating IκB. GC loading depleted the circulating pool of 25OHD3 in serum and CSF, elevated VDR mRNA and protein levels but had an inhibitory effect on CYP24A1 and VDBP expression. Vitamin D3 supplementation had an antidepressant-like effect, decreasing the immobility time and stimulating active behavior. VD3 caused a decrease in the size of the perikaryon and nucleus in CA1 hippocampal area. We found a recovery in depolarization-induced fusion of synaptic vesicles and long-term synaptic plasticity after VD3 treatment. VD3 diminished the intensity of oxidative-nitrosative stress, and suppressed the NF-κB activation. Its ameliorative effect on GC-induced neuroanatomical and behavioral abnormalities was accompanied by the 25OHD3 repletion and partial restoration of the VD3-auto/paracrine system.ConclusionGC-induced neurotoxicity and behavioral disturbances are associated with increased oxidative-nitrosative stress and impairments of VD3 metabolism. Thus, VD3 can be effective in preventing structural and functional abnormalities in the brain and behavior changes caused by long-term GC administration.
Collapse
Affiliation(s)
- Olha Lisakovska
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
- *Correspondence: Olha Lisakovska,
| | - Dmytro Labudzynskyi
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Anna Khomenko
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Dmytro Isaev
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv, Ukraine
| | - Alina Savotchenko
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv, Ukraine
| | - Ludmila Kasatkina
- Research Laboratory for Young Scientists, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Serhii Savosko
- Department of Histology and Embryology, Bogomolets National Medical University, Kyiv, Ukraine
| | - Mykola Veliky
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Ihor Shymanskyi
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
| |
Collapse
|
19
|
Manzo C, Castagna A, Ruberto C, Ruotolo G. Does a steroid dementia syndrome really exist? A brief narrative review of what the literature highlights about the relationship between glucocorticoids and cognition. GERIATRIC CARE 2023. [DOI: 10.4081/gc.2022.10975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023] Open
Abstract
Glucocorticoids (GCs) may cause cognitive impairment through complex pathways involving specific receptors. In the human brain, hippocampal CA1 neurons exhibit the highest level of GCs receptors. Even the elderly expressed these receptors. The purpose of this brief review is to concentrate on the relationship between GCs and cognition in order to discuss the effects of the so-called steroid dementia in routine clinical practice.
Collapse
|
20
|
Liu MY, Wei LL, Zhu XH, Ding HC, Liu XH, Li H, Li YY, Han Z, Li LD, Du ZW, Zhou YP, Zhang J, Meng F, Tang YL, Liu X, Wang C, Zhou QG. Prenatal stress modulates HPA axis homeostasis of offspring through dentate TERT independently of glucocorticoids receptor. Mol Psychiatry 2023; 28:1383-1395. [PMID: 36481932 PMCID: PMC10005958 DOI: 10.1038/s41380-022-01898-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 11/18/2022] [Indexed: 12/13/2022]
Abstract
In response to stressful events, the hypothalamic-pituitary-adrenal (HPA) axis is activated, and consequently glucocorticoids are released by the adrenal gland into the blood circulation. A large body of research has illustrated that excessive glucocorticoids in the hippocampus exerts negative feedback regulation of the HPA axis through glucocorticoid receptor (GR), which is critical for the homeostasis of the HPA axis. Maternal prenatal stress causes dysfunction of the HPA axis feedback mechanism in their offspring in adulthood. Here we report that telomerase reverse transcriptase (TERT) gene knockout causes hyperactivity of the HPA axis without hippocampal GR deficiency. We found that the level of TERT in the dentate gyrus (DG) of the hippocampus during the developmental stage determines the responses of the HPA axis to stressful events in adulthood through modulating the excitability of the dentate granular cells (DGCs) rather than the expression of GR. Our study also suggests that the prenatal high level of glucocorticoids exposure-induced hypomethylation at Chr13:73764526 in the first exon of mouse Tert gene accounted for TERT deficiency in the DG and HPA axis abnormality in the adult offspring. This study reveals a novel GR-independent mechanism underlying prenatal stress-associated HPA axis impairment, providing a new angle for understanding the mechanisms for maintaining HPA axis homeostasis.
Collapse
Affiliation(s)
- Meng-Ying Liu
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Department of Pharmacy, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lu-Lu Wei
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xian-Hui Zhu
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmacy, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Hua-Chen Ding
- Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Xiang-Hu Liu
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Huan Li
- School of Applied Science, Temasek Polytechnic, Singapore, Singapore.,College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuan-Yuan Li
- Department of Clinical Pharmacy, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Zhou Han
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Department of Pharmacy, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lian-Di Li
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Zi-Wei Du
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Ya-Ping Zhou
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jing Zhang
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Fan Meng
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yu-Lin Tang
- Department of Clinical Pharmacy, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao Liu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Chun Wang
- Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China.
| | - Qi-Gang Zhou
- State Key Laboratory of Reproductive Medicine, Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China. .,Department of Clinical Pharmacy, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China. .,Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China. .,The Key Center of Gene Technology Drugs of Jiangsu Province, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
21
|
Kraus KL, Nawreen N, Godale CM, Chordia AP, Packard B, LaSarge CL, Herman JP, Danzer SC. Hippocampal glucocorticoid receptors modulate status epilepticus severity. Neurobiol Dis 2023; 178:106014. [PMID: 36702319 PMCID: PMC10055427 DOI: 10.1016/j.nbd.2023.106014] [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: 11/03/2022] [Revised: 01/04/2023] [Accepted: 01/22/2023] [Indexed: 01/24/2023] Open
Abstract
Status epilepticus (SE) is a life-threatening medical emergency with significant morbidity and mortality. SE is associated with a robust and sustained increase in serum glucocorticoids, reaching concentrations sufficient to activate the dense population of glucocorticoid receptors (GRs) expressed among hippocampal excitatory neurons. Glucocorticoid exposure can increase hippocampal neuron excitability; however, whether activation of hippocampal GRs during SE exacerbates seizure severity remains unknown. To test this, a viral strategy was used to delete GRs from a subset of hippocampal excitatory neurons in adult male and female mice, producing hippocampal GR knockdown mice. Two weeks after GR knockdown, mice were challenged with the convulsant drug pilocarpine to induce SE. GR knockdown had opposing effects on early vs late seizure behaviors, with sex influencing responses. For both male and female mice, the onset of mild behavioral seizures was accelerated by GR knockdown. In contrast, GR knockdown delayed the onset of more severe convulsive seizures and death in male mice. Concordantly, GR knockdown also blunted the SE-induced rise in serum corticosterone in male mice. GR knockdown did not alter survival times or serum corticosterone in females. To assess whether loss of GR affected susceptibility to SE-induced cell death, within-animal analyses were conducted comparing local GR knockdown rates to local cell loss. GR knockdown did not affect the degree of localized neuronal loss, suggesting cell-intrinsic GR signaling neither protects nor sensitizes neurons to acute SE-induced death. Overall, the findings reveal that hippocampal GRs exert an anti-convulsant role in both males and females in the early stages of SE, followed by a switch to a pro-convulsive role for males only. Findings reveal an unexpected complexity in the interaction between hippocampal GR activation and the progression of SE.
Collapse
Affiliation(s)
- Kimberly L Kraus
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America; Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America; Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America.
| | - Nawshaba Nawreen
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America; Department of Pharmacology and Systems Physiology, University of Cincinnati School of Medicine, Cincinnati, OH, United States of America.
| | - Christin M Godale
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America; Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America.
| | - Arihant P Chordia
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America.
| | - Ben Packard
- Department of Pharmacology and Systems Physiology, University of Cincinnati School of Medicine, Cincinnati, OH, United States of America.
| | - Candi L LaSarge
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America; Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America; Department of Anesthesiology, University of Cincinnati School of Medicine, Cincinnati, OH, United States of America
| | - James P Herman
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America; Department of Anesthesiology, University of Cincinnati School of Medicine, Cincinnati, OH, United States of America.
| | - Steve C Danzer
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America; Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America; Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America; Department of Anesthesiology, University of Cincinnati School of Medicine, Cincinnati, OH, United States of America.
| |
Collapse
|
22
|
Sherman BE, Harris BB, Turk-Browne NB, Sinha R, Goldfarb EV. Hippocampal mechanisms support cortisol-induced memory enhancements. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527745. [PMID: 36798309 PMCID: PMC9934703 DOI: 10.1101/2023.02.08.527745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Stress can powerfully influence episodic memory, often enhancing memory encoding for emotionally salient information. These stress-induced memory enhancements stand at odds with demonstrations that stress and the stress-related hormone cortisol can negatively affect the hippocampus, a brain region important for episodic memory encoding. To resolve this apparent conflict and determine whether and how the hippocampus supports memory encoding under cortisol, we combined behavioral assays of associative memory, high-resolution functional magnetic resonance imaging (fMRI), and pharmacological manipulation of cortisol in a within-participant, double-blinded procedure. Hydrocortisone led to enhanced functional connectivity between hippocampal subregions, which predicted subsequent memory enhancements for emotional information. Cortisol also modified the relationship between hippocampal representations and memory: whereas hippocampal signatures of distinctiveness predicted memory under placebo, relative integration predicted memory under cortisol. Together, these data provide novel evidence that the human hippocampus contains the necessary machinery to support emotional memory enhancements under stress.
Collapse
Affiliation(s)
| | | | | | | | - Elizabeth V Goldfarb
- Department of Psychology, Yale University
- Wu Tsai Institute, Yale University
- Department of Psychiatry, Yale University
| |
Collapse
|
23
|
Wang M, Yang Y, Xu Y. Brain nuclear receptors and cardiovascular function. Cell Biosci 2023; 13:14. [PMID: 36670468 PMCID: PMC9854230 DOI: 10.1186/s13578-023-00962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Brain-heart interaction has raised up increasing attentions. Nuclear receptors (NRs) are abundantly expressed in the brain, and emerging evidence indicates that a number of these brain NRs regulate multiple aspects of cardiovascular diseases (CVDs), including hypertension, heart failure, atherosclerosis, etc. In this review, we will elaborate recent findings that have established the physiological relevance of brain NRs in the context of cardiovascular function. In addition, we will discuss the currently available evidence regarding the distinct neuronal populations that respond to brain NRs in the cardiovascular control. These findings suggest connections between cardiac control and brain dynamics through NR signaling, which may lead to novel tools for the treatment of pathological changes in the CVDs.
Collapse
Affiliation(s)
- Mengjie Wang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yongjie Yang
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
24
|
Touron E, Moulinet I, Kuhn E, Sherif S, Ourry V, Landeau B, Mézenge F, Vivien D, Klimecki OM, Poisnel G, Marchant NL, Chételat G. Depressive symptoms in cognitively unimpaired older adults are associated with lower structural and functional integrity in a frontolimbic network. Mol Psychiatry 2022; 27:5086-5095. [PMID: 36258017 PMCID: PMC9763117 DOI: 10.1038/s41380-022-01772-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 08/19/2022] [Accepted: 08/30/2022] [Indexed: 01/14/2023]
Abstract
Subclinical depressive symptoms are associated with increased risk of Alzheimer's disease (AD), but the brain mechanisms underlying this relationship are still unclear. We aimed to provide a comprehensive overview of the brain substrates of subclinical depressive symptoms in cognitively unimpaired older adults using complementary multimodal neuroimaging data. We included cognitively unimpaired older adults from the baseline data of the primary cohort Age-Well (n = 135), and from the replication cohort ADNI (n = 252). In both cohorts, subclinical depressive symptoms were assessed using the 15-item version of the Geriatric Depression Scale; based on this scale, participants were classified as having depressive symptoms (>0) or not (0). Voxel-wise between-group comparisons were performed to highlight differences in gray matter volume, glucose metabolism and amyloid deposition; as well as white matter integrity (only available in Age-Well). Age-Well participants with subclinical depressive symptoms had lower gray matter volume in the hippocampus and lower white matter integrity in the fornix and the posterior parts of the cingulum and corpus callosum, compared to participants without symptoms. Hippocampal atrophy was recovered in ADNI, where participants with subclinical depressive symptoms also showed glucose hypometabolism in the hippocampus, amygdala, precuneus/posterior cingulate cortex, medial and dorsolateral prefrontal cortex, insula, and temporoparietal cortex. Subclinical depressive symptoms were not associated with brain amyloid deposition in either cohort. Subclinical depressive symptoms in ageing are linked with neurodegeneration biomarkers in the frontolimbic network including brain areas particularly sensitive to AD. The relationship between depressive symptoms and AD may be partly underpinned by neurodegeneration in common brain regions.
Collapse
Affiliation(s)
- Edelweiss Touron
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France
| | - Inès Moulinet
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France
| | - Elizabeth Kuhn
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France
| | - Siya Sherif
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France
| | - Valentin Ourry
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France
- Unité 1077 NIMH "Neuropsychologie et Imagerie de la Mémoire Humaine," Institut National de la Santé et de la Recherche Médicale, Normandie Université, Université de Caen, PSL Université, EPHE, CHU de Caen-Normandie, GIP Cyceron, Caen, France
| | - Brigitte Landeau
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France
| | - Florence Mézenge
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France
| | - Denis Vivien
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France
- Département de Recherche Clinique, CHU de Caen-Normandie, Caen, France
| | - Olga M Klimecki
- Clinical Psychology and Behavioral Neuroscience, Faculty of Psychology, Technische Universität Dresden, 01187, Dresden, Germany
| | - Géraldine Poisnel
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France
| | | | - Gaël Chételat
- Unité 1237 PhIND "Physiopathology and Imaging of Neurological Disorders", Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Normandie Université, Université de Caen, Caen, France.
| |
Collapse
|
25
|
Cawley E, Piazza G, Das RK, Kamboj SK. A systematic review of the pharmacological modulation of autobiographical memory specificity. Front Psychol 2022; 13:1045217. [PMID: 36452391 PMCID: PMC9703074 DOI: 10.3389/fpsyg.2022.1045217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/27/2022] [Indexed: 08/31/2023] Open
Abstract
Background Over-general autobiographical memory (AM) retrieval is proposed to have a causal role in the maintenance of psychological disorders like depression and PTSD. As such, the identification of drugs that modulate AM specificity may open up new avenues of research on pharmacological modeling and treatment of psychological disorders. Aim The current review summarizes randomized, placebo-controlled studies of acute pharmacological modulation of AM specificity. Method A systematic search was conducted of studies that examined the acute effects of pharmacological interventions on AM specificity in human volunteers (healthy and clinical participants) measured using the Autobiographical Memory Test. Results Seventeen studies were identified (986 total participants), of which 16 were judged to have low risk of bias. The presence and direction of effects varied across drugs and diagnostic status of participants (clinical vs. healthy volunteers). The most commonly studied drug-hydrocortisone-produced an overall impairment in AM specificity in healthy volunteers [g = -0.28, CI (-0.53, -0.03), p = 0.03], although improvements were reported in two studies of clinical participants. In general, studies of monoamine modulators reported no effect on specificity. Conclusion Pharmacological enhancement of AM specificity is inconsistent, although monaminergic modulators show little promise in this regard. Drugs that reduce AM specificity in healthy volunteers may be useful experimental-pharmacological tools that mimic an important transdiagnostic impairment in psychological disorders. Systematic review registration PROSPERO, identifier CRD42020199076, https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42020199076.
Collapse
Affiliation(s)
- Emma Cawley
- Research Department of Clinical, Educational and Health Psychology, University College London, London, United Kingdom
| | | | | | | |
Collapse
|
26
|
Tartt AN, Mariani MB, Hen R, Mann JJ, Boldrini M. Dysregulation of adult hippocampal neuroplasticity in major depression: pathogenesis and therapeutic implications. Mol Psychiatry 2022; 27:2689-2699. [PMID: 35354926 PMCID: PMC9167750 DOI: 10.1038/s41380-022-01520-y] [Citation(s) in RCA: 192] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/22/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023]
Abstract
Major depressive disorder (MDD) was previously hypothesized to be a disease of monoamine deficiency in which low levels of monoamines in the synaptic cleft were believed to underlie depressive symptoms. More recently, however, there has been a paradigm shift toward a neuroplasticity hypothesis of depression in which downstream effects of antidepressants, such as increased neurogenesis, contribute to improvements in cognition and mood. This review takes a top-down approach to assess how changes in behavior and hippocampal-dependent circuits may be attributed to abnormalities at the molecular, structural, and synaptic level. We conclude with a discussion of how antidepressant treatments share a common effect in modulating neuroplasticity and consider outstanding questions and future perspectives.
Collapse
Affiliation(s)
| | | | - Rene Hen
- Departments of Psychiatry, Columbia University, New York, NY, USA
- Neuroscience, Columbia University, New York, NY, USA
- Pharmacology, Columbia University, New York, NY, USA
- Integrative Neuroscience, NYS Psychiatric Institute, New York, NY, USA
| | - J John Mann
- Departments of Psychiatry, Columbia University, New York, NY, USA
- Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY, USA
| | - Maura Boldrini
- Departments of Psychiatry, Columbia University, New York, NY, USA.
- Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
27
|
Kraus KL, Chordia AP, Drake AW, Herman JP, Danzer SC. Hippocampal interneurons are direct targets for circulating glucocorticoids. J Comp Neurol 2022; 530:2100-2112. [PMID: 35397117 PMCID: PMC9232959 DOI: 10.1002/cne.25322] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 11/08/2022]
Abstract
The hippocampus has become a significant target of stress research in recent years because of its role in cognitive functioning, neuropathology, and regulation of the hypothalamic-pituitary-adrenal (HPA) axis. Despite the pervasive impact of stress on psychiatric and neurological disease, many of the circuit- and cell-dependent mechanisms giving rise to the limbic regulation of the stress response remain unknown. Hippocampal excitatory neurons generally express high levels of glucocorticoid receptors (GRs) and are therefore positioned to respond directly to serum glucocorticoids. These neurons are, in turn, regulated by neighboring interneurons, subtypes of which have been shown to respond to stress exposure. However, GR expression among hippocampal interneurons is not well characterized. To determine whether key interneuron populations are direct targets for glucocorticoid action, we used two transgenic mouse lines to label parvalbumin-positive (PV+) and somatostatin-positive (SST+) interneurons. GR immunostaining of labeled interneurons was characterized within the dorsal and ventral dentate hilus, dentate cell body layer, and CA1 and CA3 stratum oriens and stratum pyramidale. While nearly all hippocampal SST+ interneurons expressed GR across all regions, GR labeling of PV+ interneurons showed considerable subregion variability. The percentage of PV+, GR+ cells was highest in the CA3 stratum pyramidale and lowest in the CA1 stratum oriens, with other regions showing intermediate levels of expression. Together, these findings indicate that, under baseline conditions, hippocampal SST+ interneurons are a ubiquitous glucocorticoid target, while only distinct populations of PV+ interneurons are direct targets. This anatomical diversity suggests functional differences in the regulation of stress-dependent hippocampal responses.
Collapse
Affiliation(s)
- Kimberly L Kraus
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Arihant P Chordia
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Austin W Drake
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - James P Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
Merabet N, Lucassen PJ, Crielaard L, Stronks K, Quax R, Sloot PMA, la Fleur SE, Nicolaou M. How exposure to chronic stress contributes to the development of type 2 diabetes: A complexity science approach. Front Neuroendocrinol 2022; 65:100972. [PMID: 34929260 DOI: 10.1016/j.yfrne.2021.100972] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/24/2021] [Accepted: 12/12/2021] [Indexed: 11/18/2022]
Abstract
Chronic stress contributes to the onset of type 2 diabetes (T2D), yet the underlying etiological mechanisms are not fully understood. Responses to stress are influenced by earlier experiences, sex, emotions and cognition, and involve a complex network of neurotransmitters and hormones, that affect multiple biological systems. In addition, the systems activated by stress can be altered by behavioral, metabolic and environmental factors. The impact of stress on metabolic health can thus be considered an emergent process, involving different types of interactions between multiple variables, that are driven by non-linear dynamics at different spatiotemporal scales. To obtain a more comprehensive picture of the links between chronic stress and T2D, we followed a complexity science approach to build a causal loop diagram (CLD) connecting the various mediators and processes involved in stress responses relevant for T2D pathogenesis. This CLD could help develop novel computational models and formulate new hypotheses regarding disease etiology.
Collapse
Affiliation(s)
- Nadège Merabet
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health Research Institute, Meibergdreef 9, Amsterdam, the Netherlands; Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands
| | - Paul J Lucassen
- Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Brain Plasticity Group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Loes Crielaard
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health Research Institute, Meibergdreef 9, Amsterdam, the Netherlands; Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands
| | - Karien Stronks
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health Research Institute, Meibergdreef 9, Amsterdam, the Netherlands; Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands
| | - Rick Quax
- Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Computational Science Lab, University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Peter M A Sloot
- Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Computational Science Lab, University of Amsterdam, Amsterdam 1098 XH, the Netherlands; National Centre of Cognitive Research, ITMO University, St. Petersburg, Russian Federation
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism & Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, the Netherlands.
| | - Mary Nicolaou
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health Research Institute, Meibergdreef 9, Amsterdam, the Netherlands; Institute for Advanced Study, University of Amsterdam, Amsterdam 1012 GC, the Netherlands; Centre for Urban Mental Health, University of Amsterdam, Amsterdam 1012 GC, the Netherlands.
| |
Collapse
|
29
|
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: 30] [Impact Index Per Article: 10.0] [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
|
30
|
Hippocampal neuropathology in suicide: Gaps in our knowledge and opportunities for a breakthrough. Neurosci Biobehav Rev 2021; 132:542-552. [PMID: 34906612 DOI: 10.1016/j.neubiorev.2021.12.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 12/03/2021] [Accepted: 12/10/2021] [Indexed: 01/27/2023]
Abstract
Suicide is a major global hazard. There is a need for increasing suicide awareness and effective and evidence-based interventions, targeting both suicidal ideation and conduct. However, anti-suicide pharmacological effects are unsatisfactory. The human hippocampus is vulnerable to neuropsychiatric damages and subsequently releases psychobiological signals. Human hippocampal studies of suicide completers have shown mechanistic changes in neurobiology, which, however, could not reflect the neuropathological 'fingerprints' of fatal suicide ideations and suicide attempts. In this review, we provide several leading theories of suicide, including the serotoninergic system, Wnt pathway and brain-derived neurotrophic factor/tropomyosin receptor kinase B signalling, and discuss the evidence for their roles in suicide and treatment. Moreover, the cognitive dysfunctions associated with suicide risk are discussed, as well as the novel evidence on cognitive therapies that decrease suicidal ideation. We highlight the need to apply multi-omics techniques (including single-nucleus RNA sequencing and mass spectrometry histochemistry) on hippocampal samples from donors who died by suicide or legal euthanasia, to clarify the aetiology of suicide and propose novel therapeutic strategies.
Collapse
|
31
|
Espina JEC, Bagamasbad PD. Synergistic gene regulation by thyroid hormone and glucocorticoid in the hippocampus. VITAMINS AND HORMONES 2021; 118:35-81. [PMID: 35180933 DOI: 10.1016/bs.vh.2021.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The hippocampus is considered the center for learning and memory in the brain, and its development and function is greatly affected by the thyroid and stress axes. Thyroid hormone (TH) and glucocorticoids (GC) are known to have a synergistic effect on developmental programs across several vertebrate species, and their effects on hippocampal structure and function are well-documented. However, there are few studies that focus on the processes and genes that are cooperatively regulated by the two hormone axes. Cross-regulation of the thyroid and stress axes in the hippocampus occurs on multiple levels such that TH can regulate the expression of the GC receptor (GR) while GC can modulate tissue sensitivity to TH by controlling the expression of TH receptor (TR) and enzymes involved in TH biosynthesis. Thyroid hormone and GC are also known to synergistically regulate the transcription of genes associated with neuronal function and development. Synergistic gene regulation by TH and GC may occur through the direct, cooperative action of TR and GR on common target genes, or by indirect mechanisms involving gene regulatory cascades activated by TR and GR. In this chapter, we describe the known physiological effects and underlying molecular mechanisms of TH and GC synergistic gene regulation in the hippocampus.
Collapse
Affiliation(s)
- Jose Ezekiel C Espina
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines
| | - Pia D Bagamasbad
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City, Philippines.
| |
Collapse
|
32
|
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
|
33
|
Medina J, De Guzman RM, Workman JL. Lactation is not required for maintaining maternal care and active coping responses in chronically stressed postpartum rats: Interactions between nursing demand and chronic variable stress. Horm Behav 2021; 136:105035. [PMID: 34488064 DOI: 10.1016/j.yhbeh.2021.105035] [Citation(s) in RCA: 4] [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: 01/27/2021] [Revised: 06/11/2021] [Accepted: 07/04/2021] [Indexed: 10/20/2022]
Abstract
Women who do not breastfeed or discontinue breastfeeding early are more likely to develop postpartum depression (PPD) and stress is a significant risk factor for depression, including PPD. Using a rat model, we investigated whether the absence of nursing would increase the susceptibility to chronic stress-related behavioral and neural changes during the postpartum period. Adult female rats underwent thelectomy (thel; removal of teats), sham surgery, or no surgery (control) and were paired with males for breeding. All litters were rotated twice daily until postpartum day (PD) 26. Sham rats served as surrogates for thel litters, yielding a higher nursing demand for sham rats. Concurrently, rats received either no stress or chronic variable stress until PD 25. Rats were observed for maternal behaviors and tested in a series of tasks including open field, sucrose preference, and forced swim. We used immunohistochemistry (IHC) for doublecortin (DCX; to label immature neurons) or for mineralocorticoid receptor (MR). Contrary to our expectations, non-nursing thel rats were resistant to the effects of stress in all dependent measures. Our data indicate that even in chronic adverse conditions, nursing is not required for maintaining stable care to offspring or active coping responses in an acutely stressful task. We discuss the possible role of offspring contact and consider future directions for biomedical and clinical research. In rats with high nursing demand, however, chronic stress increased immobility, hippocampal neurogenesis, and MR expression (largely in opposition to the effects of stress in rats with typical nursing demand). We discuss these patterns in the context of energetics and allostatic load. This research highlights the complexity in relationships between stress, nursing, and neurobehavioral outcomes in the postpartum period and underscores the need for additional biomedical and clinical research geared toward optimizing treatments and interventions for women with PPD, regardless of breastfeeding status. SIGNIFICANCE STATEMENT: The goal of this research was to determine how the absence of nursing and higher nursing demand impact stress-coping behaviors and neural changes associated with chronic stress in order to disentangle the complex interplay of factors that contribute to psychological illness during the postpartum period.
Collapse
Affiliation(s)
- Joanna Medina
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America
| | - Rose M De Guzman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America
| | - Joanna L Workman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America; Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America.
| |
Collapse
|
34
|
Chae WR, Metz S, Pantazidis P, Dziobek I, Hellmann-Regen J, Wingenfeld K, Otte C. Effects of glucocorticoid and noradrenergic activity on implicit and explicit facial emotion recognition in healthy young men. Stress 2021; 24:1050-1056. [PMID: 33860721 DOI: 10.1080/10253890.2021.1908255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The ability to recognize emotions from facial expressions is crucial for social interaction. Only few studies have examined the effect of stress hormones on facial emotion recognition, although stressful events affect social interactions on a daily basis. Those studies that examined facial emotion recognition mostly used explicit prompts to trigger consciously controlled processing. However, facial emotions are processed mainly implicitly in real life. Therefore, we investigated separate and combined effects of noradrenergic and glucocorticoid stimulation on implicit and explicit facial emotion recognition. One hundred and four healthy men (mean age = 24.1 years ±SD 3.5) underwent the Face Puzzle task to test implicit and explicit facial emotion recognition after receiving either 10 mg hydrocortisone or 10 mg yohimbine (an alpha 2-adrenergic receptor antagonist that increases noradrenergic activity) or 10 mg hydrocortisone/10 mg yohimbine combined or placebo. Salivary cortisol and salivary alpha amylase (sAA) were measured during the experiment. Compared to the placebo condition hydrocortisone significantly increased salivary cortisol and yohimbine significantly increased sAA. Participants were better and faster in explicit than in implicit facial emotion recognition. However, there was no effect of separate and combined noradrenergic and glucocorticoid stimulation on implicit and explicit facial emotion recognition performance compared to placebo. Our results do not support an essential role of the glucocorticoid and noradrenergic system in FER in young healthy men.
Collapse
Affiliation(s)
- Woo Ri Chae
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Sophie Metz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Pierre Pantazidis
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Isabel Dziobek
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julian Hellmann-Regen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Katja Wingenfeld
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Christian Otte
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
| |
Collapse
|
35
|
Yong SJ, Yong MH, Teoh SL, Soga T, Parhar I, Chew J, Lim WL. The Hippocampal Vulnerability to Herpes Simplex Virus Type I Infection: Relevance to Alzheimer's Disease and Memory Impairment. Front Cell Neurosci 2021; 15:695738. [PMID: 34483839 PMCID: PMC8414573 DOI: 10.3389/fncel.2021.695738] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) as a possible infectious etiology in Alzheimer’s disease (AD) has been proposed since the 1980s. The accumulating research thus far continues to support the association and a possible causal role of HSV-1 in the development of AD. HSV-1 has been shown to induce neuropathological and behavioral changes of AD, such as amyloid-beta accumulation, tau hyperphosphorylation, as well as memory and learning impairments in experimental settings. However, a neuroanatomical standpoint of HSV-1 tropism in the brain has not been emphasized in detail. In this review, we propose that the hippocampal vulnerability to HSV-1 infection plays a part in the development of AD and amnestic mild cognitive impairment (aMCI). Henceforth, this review draws on human studies to bridge HSV-1 to hippocampal-related brain disorders, namely AD and aMCI/MCI. Next, experimental models and clinical observations supporting the neurotropism or predilection of HSV-1 to infect the hippocampus are examined. Following this, factors and mechanisms predisposing the hippocampus to HSV-1 infection are discussed. In brief, the hippocampus has high levels of viral cellular receptors, neural stem or progenitor cells (NSCs/NPCs), glucocorticoid receptors (GRs) and amyloid precursor protein (APP) that support HSV-1 infectivity, as well as inadequate antiviral immunity against HSV-1. Currently, the established diseases HSV-1 causes are mucocutaneous lesions and encephalitis; however, this review revises that HSV-1 may also induce and/or contribute to hippocampal-related brain disorders, especially AD and aMCI/MCI.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Min Hooi Yong
- Department of Psychology, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia.,Aging Health and Well-being Research Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Tomoko Soga
- Jeffrey Cheah School of Medicine and Health Sciences, Brain Research Institute Monash Sunway, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ishwar Parhar
- Jeffrey Cheah School of Medicine and Health Sciences, Brain Research Institute Monash Sunway, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jactty Chew
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia.,Aging Health and Well-being Research Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| |
Collapse
|
36
|
Peroxiredoxin 6 Knockout Mice Demonstrate Anxiety Behavior and Attenuated Contextual Fear Memory after Receiving Acute Immobilization Stress. Antioxidants (Basel) 2021; 10:antiox10091416. [PMID: 34573048 PMCID: PMC8466988 DOI: 10.3390/antiox10091416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Stress can elicit glucocorticoid release to promote coping mechanisms and influence learning and memory performance. Individual memory performance varies in response to stress, and the underlying mechanism is not clear yet. Peroxiredoxin 6 (PRDX6) is a multifunctional enzyme participating in both physiological and pathological conditions. Several studies have demonstrated the correlation between PRDX6 expression level and stress-related disorders. Our recent finding indicates that lack of the Prdx6 gene leads to enhanced fear memory. However, it is unknown whether PRDX6 is involved in changes in anxiety response and memory performance upon stress. The present study reveals that hippocampal PRDX6 level is downregulated 30 min after acute immobilization stress (AIS) and trace fear conditioning (TFC). In human retinal pigment epithelium (ARPE-19) cells, the PRDX6 expression level decreases after being treated with stress hormone corticosterone. Lack of PRDX6 caused elevated basal H2O2 levels in the hippocampus, basolateral amygdala, and medial prefrontal cortex, brain regions involved in anxiety response and fear memory formation. Additionally, this H2O2 level was still high in the medial prefrontal cortex of the knockout mice under AIS. Anxiety behavior of Prdx6-/- mice was enhanced after immobilization for 30 min. After exposure to AIS before a contextual test, Prdx6-/- mice displayed a contextual fear memory deficit. Our results showed that the memory performance of Prdx6-/- mice was impaired when responding to AIS, accompanied by dysregulated H2O2 levels. The present study helps better understand the function of PRDX6 in memory performance after acute stress.
Collapse
|
37
|
Dream Recall/Affect and the Hypothalamic-Pituitary-Adrenal Axis. Clocks Sleep 2021; 3:403-408. [PMID: 34449575 PMCID: PMC8395463 DOI: 10.3390/clockssleep3030027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/08/2021] [Accepted: 07/16/2021] [Indexed: 12/02/2022] Open
Abstract
In this concise review, we present an overview of research on dream recall/affect and of the hypothalamic–pituitary–adrenal (HPA) axis, discussing caveats regarding the action of hormones of the HPA axis (mainly cortisol and its free form, cortisol-binding globulin and glucocorticoid receptors). We present results of studies regarding dream recall/affect and the HPA axis under physiological (such as waking) or pathological conditions (such as in Cushing’s syndrome or stressful situations). Finally, we try to integrate the effect of the current COVID-19 situation with dream recall/affect vis-à-vis the HPA axis.
Collapse
|
38
|
Moog NK, Nolvi S, Kleih TS, Styner M, Gilmore JH, Rasmussen JM, Heim CM, Entringer S, Wadhwa PD, Buss C. Prospective association of maternal psychosocial stress in pregnancy with newborn hippocampal volume and implications for infant social-emotional development. Neurobiol Stress 2021; 15:100368. [PMID: 34355050 PMCID: PMC8319845 DOI: 10.1016/j.ynstr.2021.100368] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/30/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
Maternal psychosocial stress during pregnancy can impact the developing fetal brain and influence offspring mental health. In this context, animal studies have identified the hippocampus and amygdala as key brain regions of interest, however, evidence in humans is sparse. We, therefore, examined the associations between maternal prenatal psychosocial stress, newborn hippocampal and amygdala volumes, and child social-emotional development. In a sample of 86 mother-child dyads, maternal perceived stress was assessed serially in early, mid and late pregnancy. Following birth, newborn (aged 5–64 postnatal days, mean: 25.8 ± 12.9) hippocampal and amygdala volume was assessed using structural magnetic resonance imaging. Infant social-emotional developmental milestones were assessed at 6- and 12-months age using the Bayley-III. After adjusting for covariates, maternal perceived stress during pregnancy was inversely associated with newborn left hippocampal volume (β = −0.26, p = .019), but not with right hippocampal (β = −0.170, p = .121) or bilateral amygdala volumes (ps > .5). Furthermore, newborn left hippocampal volume was positively associated with infant social-emotional development across the first year of postnatal life (B = 0.01, p = .011). Maternal perceived stress was indirectly associated with infant social-emotional development via newborn left hippocampal volume (B = −0.34, 95% CIBC [-0.97, −0.01]), suggesting mediation. This study provides prospective evidence in humans linking maternal psychosocial stress in pregnancy with newborn hippocampal volume and subsequent infant social-emotional development across the first year of life. These findings highlight the importance of maternal psychosocial state during pregnancy as a target amenable to interventions to prevent or attenuate its potentially unfavorable neural and behavioral consequences in the offspring. Maternal perceived stress predicted smaller neonatal left hippocampal volume (HCV). Neonatal left HCV was positively associated with infant social-emotional function. Variation in HCV may mediate maternal stress-related effects on child mental health.
Collapse
Affiliation(s)
- Nora K Moog
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Medical Psychology, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Saara Nolvi
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Medical Psychology, Augustenburger Platz 1, 13353, Berlin, Germany.,Turku Institute for Advanced Studies, Department of Psychology and Speech-Language Pathology, University of Turku, Finland
| | - Theresa S Kleih
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Medical Psychology, Augustenburger Platz 1, 13353, Berlin, Germany.,Institute of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin Styner
- Departments of Psychiatry and Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jerod M Rasmussen
- Development, Health, and Disease Research Program, Departments of Pediatrics, Psychiatry and Human Behavior, Obstetrics and Gynecology, and Epidemiology, University of California, Irvine, School of Medicine, Irvine, CA, USA
| | - Christine M Heim
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Medical Psychology, Augustenburger Platz 1, 13353, Berlin, Germany.,Department of Biobehavioral Health, Pennsylvania State University, College of Health and Human Development, University Park, PA, USA
| | - Sonja Entringer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Medical Psychology, Augustenburger Platz 1, 13353, Berlin, Germany.,Development, Health, and Disease Research Program, Departments of Pediatrics, Psychiatry and Human Behavior, Obstetrics and Gynecology, and Epidemiology, University of California, Irvine, School of Medicine, Irvine, CA, USA.,Department of Pediatrics, University of California, Irvine, School of Medicine, Orange, CA, USA
| | - Pathik D Wadhwa
- Development, Health, and Disease Research Program, Departments of Pediatrics, Psychiatry and Human Behavior, Obstetrics and Gynecology, and Epidemiology, University of California, Irvine, School of Medicine, Irvine, CA, USA.,Department of Pediatrics, University of California, Irvine, School of Medicine, Orange, CA, USA.,Departments of Psychiatry and Human Behavior, Obstetrics and Gynecology, and Epidemiology, University of California, Irvine, School of Medicine, Orange, CA, USA
| | - Claudia Buss
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Medical Psychology, Augustenburger Platz 1, 13353, Berlin, Germany.,Development, Health, and Disease Research Program, Departments of Pediatrics, Psychiatry and Human Behavior, Obstetrics and Gynecology, and Epidemiology, University of California, Irvine, School of Medicine, Irvine, CA, USA.,Department of Pediatrics, University of California, Irvine, School of Medicine, Orange, CA, USA
| |
Collapse
|
39
|
Mukku SSR, Dahale AB, Muniswamy NR, Muliyala KP, Sivakumar PT, Varghese M. Geriatric Depression and Cognitive Impairment-An Update. Indian J Psychol Med 2021; 43:286-293. [PMID: 34385720 PMCID: PMC8327864 DOI: 10.1177/0253717620981556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Depression and cognitive impairment often coexist in older adults. The relation between depression and cognitive impairment is complex. The objective of this article is to review recent literature on cognitive impairment in older adults with depression and provide clinicians an update. METHODS We searched PubMed, Google Scholar, Science Direct, and Psych Info for the articles published in the English language related to late-life depression (LLD)/geriatric depression and cognitive impairment. We considered original research articles, relevant systematic reviews, chapters, and important conceptual articles published in the last 9 years (2011-2019). We selected relevant articles for this narrative review. CONCLUSION The concept pseudodementia, indicating depression with cognitive impairment mimicking dementia, is now seen only as a historical concept. The current literature strongly agrees with fact that cognitive deficits often exist in LLD. The cognitive deficits in depression were initially seen as trait marker; however, some recent studies suggest that cognitive deficits persist even in the remission phase. There is heterogeneity among the studies in terms of the nature of the cognitive deficits, but higher number of studies reported impairment in attention and executive function. LLD with cognitive deficits is at a higher risk of progression to dementia. In older adults, depression with cognitive impairments requires a comprehensive evaluation. Electroencephalography, event-related potentials, fluorodeoxyglucose-positron emission tomography, amyloid positron emission tomography, and CSF amyloid will supplement clinical evaluation in differentiating functional depressive disorder with cognitive impairment from depression with an underlying degenerative condition.
Collapse
Affiliation(s)
- Shiva Shanker Reddy Mukku
- Geriatric Clinic and Services, Dept. of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Ajit Bhalchandra Dahale
- Dept. of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | | | - Krishna Prasad Muliyala
- Dept. of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Palanimuthu Thangaraju Sivakumar
- Geriatric Clinic and Services, Dept. of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Mathew Varghese
- Geriatric Clinic and Services, Dept. of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| |
Collapse
|
40
|
Mohammadi S, Zandi M, Dousti Kataj P, Karimi Zandi L. Chronic stress and Alzheimer's disease. Biotechnol Appl Biochem 2021; 69:1451-1458. [PMID: 34152660 DOI: 10.1002/bab.2216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022]
Abstract
Stress is a key factor in the development and progress of diseases. In neurodegenerative conditions, stress management can play an important role in maintaining the quality of life and the capacity to improve. Neurodegenerative diseases, including Alzheimer's disease, cause the motor and cognitive malfunctions that are spontaneously stressful and also can disturb the neural circuits that promote stress responses. The interruption of those circuits leads to aggressive and inappropriate behavior. In addition, stress contributes to illness and may exacerbate symptoms. In this review, we present stress-activated neural pathways involved in Alzheimer's disease from a clinical and experimental point of view, as well as supportive drugs and therapies.
Collapse
Affiliation(s)
- Shima Mohammadi
- Neuroscience and Addiction Studies Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Parviz Dousti Kataj
- Neuroscience and Addiction Studies Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Karimi Zandi
- Neuroscience and Addiction Studies Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Langreck C, Wauson E, Nerland D, Lamb B, Folkerts T, Winter L, Lu E, Tague S, McCarson KE, Ploski JE, Banasr M, Duman RS, Roland MM, Babich V, Di Sole F, Duric V. Hippocampal mitogen-activated protein kinase phosphatase-1 regulates behavioral and systemic effects of chronic corticosterone administration. Biochem Pharmacol 2021; 190:114617. [PMID: 34023293 DOI: 10.1016/j.bcp.2021.114617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022]
Abstract
Clinical reports indicate a bidirectional relationship between mental illness and chronic systemic diseases. However, brain mechanisms linking chronic stress and development of mood disorders to accompanying peripheral organ dysfunction are still not well characterized in animal models. In the current study, we investigated whether activation of hippocampal mitogen-activated protein kinase phosphatase-1 (MKP-1), a key factor in depression pathophysiology, also acts as a mediator of systemic effects of stress. First, we demonstrated that treatment with the glucocorticoid receptor (GR) agonist dexamethasone or acute restraint stress (ARS) significantly increased Mkp-1 mRNA levels within the rat hippocampus. Conversely, administration of the GR antagonist mifepristone 30 min before ARS produced a partial blockade of Mkp-1 upregulation, suggesting that stress activates MKP-1, at least in part, through upstream GR signaling. Chronic corticosterone (CORT) administration evoked comparable increases in hippocampal MKP-1 protein levels and produced a robust increase in behavioral emotionality. In addition to behavioral deficits, chronic CORT treatment also produced systemic pathophysiological effects. Elevated levels of renal inflammation protein markers (NGAL and IL18) were observed suggesting tissue damage and early kidney impairment. In a rescue experiment, the effects of CORT on development of depressive-like behaviors and increased NGAL and IL18 protein levels in the kidney were blocked by CRISPR-mediated knockdown of hippocampal Mkp-1 prior to CORT exposure. In sum, these findings further demonstrate that MKP-1 is necessary for development of enhanced behavioral emotionality, while also suggesting a role in stress mechanisms linking brain dysfunction and systemic illness such as kidney disease.
Collapse
Affiliation(s)
- Cory Langreck
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States; Department of Pharmacology, Columbia University, New York, NY 10032, United States
| | - Eric Wauson
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States
| | - Dakota Nerland
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States
| | - Brad Lamb
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States
| | - Tyler Folkerts
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States
| | - Lori Winter
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States
| | - Eileen Lu
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States
| | - Sarah Tague
- Smith Intellectual & Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Kenneth E McCarson
- Smith Intellectual & Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS 66160, United States; Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Jonathan E Ploski
- School of Behavioral and Brain Sciences and Department of Molecular and Cell Biology, The University of Texas at Dallas, Dallas, TX 75080, United States
| | - Mounira Banasr
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada; Department of Psychiatry, Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Ronald S Duman
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06508, United States
| | - Miranda M Roland
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States
| | - Victor Babich
- School of Liberal Arts and Sciences, Mercy College of Health Sciences, Des Moines, IA 50312, United States
| | - Francesca Di Sole
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States
| | - Vanja Duric
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, United States.
| |
Collapse
|
42
|
Exposure to 2.45 GHz Radiation Triggers Changes in HSP-70, Glucocorticoid Receptors and GFAP Biomarkers in Rat Brain. Int J Mol Sci 2021; 22:ijms22105103. [PMID: 34065959 PMCID: PMC8151023 DOI: 10.3390/ijms22105103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/23/2021] [Accepted: 05/10/2021] [Indexed: 01/27/2023] Open
Abstract
Brain tissue may be especially sensitive to electromagnetic phenomena provoking signs of neural stress in cerebral activity. Fifty-four adult female Sprague-Dawley rats underwent ELISA and immunohistochemistry testing of four relevant anatomical areas of the cerebrum to measure biomarkers indicating induction of heat shock protein 70 (HSP-70), glucocorticoid receptors (GCR) or glial fibrillary acidic protein (GFAP) after single or repeated exposure to 2.45 GHz radiation in the experimental set-up. Neither radiation regime caused tissue heating, so thermal effects can be ruled out. A progressive decrease in GCR and HSP-70 was observed after acute or repeated irradiation in the somatosensory cortex, hypothalamus and hippocampus. In the limbic cortex; however, values for both biomarkers were significantly higher after repeated exposure to irradiation when compared to control animals. GFAP values in brain tissue after irradiation were not significantly different or were even lower than those of nonirradiated animals in all brain regions studied. Our results suggest that repeated exposure to 2.45 GHz elicited GCR/HSP-70 dysregulation in the brain, triggering a state of stress that could decrease tissue anti-inflammatory action without favoring glial proliferation and make the nervous system more vulnerable.
Collapse
|
43
|
Noureddine LM, Trédan O, Hussein N, Badran B, Le Romancer M, Poulard C. Glucocorticoid Receptor: A Multifaceted Actor in Breast Cancer. Int J Mol Sci 2021; 22:ijms22094446. [PMID: 33923160 PMCID: PMC8123001 DOI: 10.3390/ijms22094446] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is one of the most common cancers in women worldwide. Even though the role of estrogen receptor alpha (ERα) is extensively documented in the development of breast tumors, other members of the nuclear receptor family have emerged as important players. Synthetic glucocorticoids (GCs) such as dexamethasone (dex) are commonly used in BC for their antiemetic, anti-inflammatory, as well as energy and appetite stimulating properties, and to manage the side effects of chemotherapy. However, dex triggers different effects depending on the BC subtype. The glucocorticoid receptor (GR) is also an important marker in BC, as high GR expression is correlated with a poor and good prognosis in ERα-negative and ERα-positive BCs, respectively. Indeed, though it drives the expression of pro-tumorigenic genes in ERα-negative BCs and is involved in resistance to chemotherapy and metastasis formation, dex inhibits estrogen-mediated cell proliferation in ERα-positive BCs. Recently, a new natural ligand for GR called OCDO was identified. OCDO is a cholesterol metabolite with oncogenic properties, triggering mammary cell proliferation in vitro and in vivo. In this review, we summarize recent data on GR signaling and its involvement in tumoral breast tissue, via its different ligands.
Collapse
Affiliation(s)
- Lara Malik Noureddine
- Université de Lyon, F-69000 Lyon, France; (L.M.N.); (O.T.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences, Lebanese University, Hadat-Beirut 90656, Lebanon; (N.H.); (B.B.)
| | - Olivier Trédan
- Université de Lyon, F-69000 Lyon, France; (L.M.N.); (O.T.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Centre Leon Bérard, Oncology Department, F-69000 Lyon, France
| | - Nader Hussein
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences, Lebanese University, Hadat-Beirut 90656, Lebanon; (N.H.); (B.B.)
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences, Lebanese University, Hadat-Beirut 90656, Lebanon; (N.H.); (B.B.)
| | - Muriel Le Romancer
- Université de Lyon, F-69000 Lyon, France; (L.M.N.); (O.T.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Coralie Poulard
- Université de Lyon, F-69000 Lyon, France; (L.M.N.); (O.T.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Correspondence: ; Tel.: +33-478-786-663; Fax: +33-478-782-720
| |
Collapse
|
44
|
Bongartz H, Seiß EA, Bock J, Schaper F. Glucocorticoids attenuate interleukin-6-induced c-Fos and Egr1 expression and impair neuritogenesis in PC12 cells. J Neurochem 2021; 157:532-549. [PMID: 33454999 DOI: 10.1111/jnc.15305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 01/15/2023]
Abstract
Interleukin-6 (IL-6) is a cytokine primarily known for immune regulation. There is also growing evidence that IL-6 triggers neurogenesis and impacts neural development, both life-long occurring processes that can be impaired by early-life and adult stress. Stress induces the release of glucocorticoids by activation of the hypothalamic-pituitary-adrenal (HPA) axis. On the cellular level, glucocorticoids act via the ubiquitously expressed glucocorticoid receptor. Thus, we aimed to elucidate whether glucocorticoids affect IL-6-induced neural development. Here, we show that IL-6 signalling induces neurite outgrowth in adrenal pheochromocytoma PC12 cells in a mitogen-activated protein kinase (MAPK) pathway-dependent manner, since neurite outgrowth was diminished upon Mek-inhibitor treatment. Using quantitative biochemical approaches, such as qRT-PCR analysis of Hyper-IL-6 treated PC12 cells, we show that neurite outgrowth induced by IL-6 signalling is accompanied by early and transient MAPK-dependent mRNA expression of immediate early genes coding for proteins such as early growth response protein 1 (Egr1) and c-Fos. This correlates with reduced proliferation and prolonged G0/G1 cell cycle arrest as determined by monitoring the cellular DNA content using flow cytometry. These results indicate for IL-6 signalling-induced neural differentiation. Interestingly, the glucocorticoid Dexamethasone impairs early IL-6 signalling-induced mRNA expression of c-Fos and Egr1 and restrains neurite outgrowth. Impaired Egr1 and c-Fos expression in neural development is implicated in the aetiology of neuropathologies. Thus, it appears likely that stress-induced release of glucocorticoids, as well as therapeutically administered glucocorticoids, contribute to the development of neuropathologies by reducing the expression of Egr1 and c-Fos, and by restraining IL-6-dependent neural differentiation.
Collapse
Affiliation(s)
- Hannes Bongartz
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Magdeburg, Germany
| | - Elena Anne Seiß
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Magdeburg, Germany
| | - Jörg Bock
- Institute of Biology, PG "Epigenetics and Structural Plasticity", Otto-von-Guericke University, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke University, Magdeburg, Germany
| | - Fred Schaper
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Magdeburg, Germany.,Center for Dynamic Systems: Systems Engineering (CDS), Otto-von-Guericke University, Magdeburg, Germany.,Magdeburg Center for Systems Biology (MACS), Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
45
|
Manzo C, Serra-Mestres J, Isetta M, Castagna A. Could COVID-19 anosmia and olfactory dysfunction trigger an increased risk of future dementia in patients with ApoE4? Med Hypotheses 2021; 147:110479. [PMID: 33422806 PMCID: PMC7785277 DOI: 10.1016/j.mehy.2020.110479] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022]
Abstract
The association of the coronavirus disease 2019 (COVID-19) with significant neurological and neuropsychiatric complications has been increasingly reported, both during the acute illness and in its aftermath. However, due to the short duration of patient follow up until now, it is not clear whether this infection will be associated with longer-term neurological and/or neuropsychiatric sequelae. In particular, the question of whether COVID-19 will be associated with an increased risk and rate of future dementia remains open and subject to speculation. During the course of the COVID-19 pandemic, an increasing number of patients have reported sudden anosmia or other olfactory dysfunction as concurrent symptoms. The possibility that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may reach the brain via the olfactory nerve or an upper nasal trancribrial route is an interesting working hypothesis. Among the identified genetic risk factors for Late-onset Alzheimer’s disease (LOAD), Apo E4 is one of the strongest and most frequent. People carrying one or two copies of the e4 allele of Apo E4 have significant odor recognition deficits in comparison to those not carrying this haplotype. The hypothesis invoked in this paper is that anosmia/olfactory dysfunctions induced by SARS-CoV-2 may cause an increased a risk of future neurodegenerative dementia in ApoE4 carriers, and that this risk would be higher than in Apo E4 carriers affected by anosmia not induced by SARS-CoV-2. This would be associated with virus-induced chronic modifications in the central nervous system. It is proposed that COVID-19 patients with anosmia and no other serious symptoms should be followed up as part of specifically designed and approved studies in order to identify the early stages of dementia (especially LOAD and Dementia with Lewy Bodies), thereby improving our knowledge of the mechanisms involved in pre-cognitive stages of neurodegenerative dementia and making best use of any available therapies. This latter opportunity is unique and should not be lost.
Collapse
Affiliation(s)
- Ciro Manzo
- Azienda Sanitaria Locale Napoli 3 sud, Internal and Geriatric Medicine Department, Center for Cognitive Disorders and Dementia, Health district no.51 - Pomigliano d'Arco Naples, Italy.
| | - Jordi Serra-Mestres
- Department of Old Age Psychiatry, Central and North West London NHS Foundation Trust, London, UK.
| | - Marco Isetta
- Library and Knowldege Services, Central and North West London NHS Foundation Trust, London, UK.
| | - Alberto Castagna
- Geriatric Medicine Department, Azienda Sanitaria Provinciale Catanzaro, Fragility Outpatient Clinic, Casa della Salute di Chiaravalle Centrale, Chiaravalle Catanzaro, Italy.
| |
Collapse
|
46
|
Kalsbeek A, Buijs RM. Organization of the neuroendocrine and autonomic hypothalamic paraventricular nucleus. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:45-63. [PMID: 34225948 DOI: 10.1016/b978-0-12-820107-7.00004-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A major function of the nervous system is to maintain a relatively constant internal environment. The distinction between our external environment (i.e., the environment that we live in and that is subject to major changes, such as temperature, humidity, and food availability) and our internal environment (i.e., the environment formed by the fluids surrounding our bodily tissues and that has a very stable composition) was pointed out in 1878 by Claude Bernard (1814-1878). Later on, it was indicated by Walter Cannon (1871-1945) that the internal environment is not really constant, but rather shows limited variability. Cannon named the mechanism maintaining this limited variability homeostasis. Claude Bernard envisioned that, for optimal health, all physiologic processes in the body needed to maintain homeostasis and should be in perfect harmony with each other. This is illustrated by the fact that, for instance, during the sleep-wake cycle important elements of our physiology such as body temperature, circulating glucose, and cortisol levels show important variations but are in perfect synchrony with each other. These variations are driven by the biologic clock in interaction with hypothalamic target areas, among which is the paraventricular nucleus of the hypothalamus (PVN), a core brain structure that controls the neuroendocrine and autonomic nervous systems and thus is key for integrating central and peripheral information and implementing homeostasis. This chapter focuses on the anatomic connections between the biologic clock and the PVN to modulate homeostasis according to the daily sleep-wake rhythm. Experimental studies have revealed a highly specialized organization of the connections between the clock neurons and neuroendocrine system as well as preautonomic neurons in the PVN. These complex connections ensure a logical coordination between behavioral, endocrine, and metabolic functions that helps the organism maintain homeostasis throughout the day.
Collapse
Affiliation(s)
- Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (Amsterdam UMC), University of Amsterdam, Amsterdam, The Netherlands; Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
| | - Ruud M Buijs
- Hypothalamic Integration Mechanisms Laboratory, Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| |
Collapse
|
47
|
Watermeyer T, Robb C, Gregory S, Udeh-Momoh C. Therapeutic implications of hypothalamic-pituitaryadrenal-axis modulation in Alzheimer's disease: A narrative review of pharmacological and lifestyle interventions. Front Neuroendocrinol 2021; 60:100877. [PMID: 33045258 DOI: 10.1016/j.yfrne.2020.100877] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
With disease-modifying treatments for Alzheimer's disease (AD) still elusive, the search for alternative intervention strategies has intensified. Growing evidence suggests that dysfunction in hypothalamic-pituitaryadrenal-axis (HPAA) activity may contribute to the development of AD pathology. The HPAA, may therefore offer a novel target for therapeutic action. This review summarises and critically evaluates animal and human studies investigating the effects of pharmacological and non-pharmacological intervention on HPAA modulation alongside cognitive performance. The interventions discussed include glucocorticoid receptor antagonists and 11β-hydroxysteroid dehydrogenase inhibitors as well as lifestyle treatments such as physical activity, diet, sleep and contemplative practices. Pharmacological HPAA modulators improve pathology and cognitive deficit in animal AD models, but human pharmacological trials are yet to provide definitive support for such benefits. Lifestyle interventions may offer promising strategies for HPAA modification and cognitive health, but several methodological caveats across these studies were identified. Directions for future research in AD studies are proposed.
Collapse
Affiliation(s)
- Tamlyn Watermeyer
- Department of Psychology, Faculty of Health and Life Sciences, Northumbria University, Newcastle, UK; Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Catherine Robb
- Ageing Epidemiology Research Unit, School of Public Health, Faculty of Medicine, The Imperial College of Science, Technology and Medicine, London, UK
| | - Sarah Gregory
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Chinedu Udeh-Momoh
- Ageing Epidemiology Research Unit, School of Public Health, Faculty of Medicine, The Imperial College of Science, Technology and Medicine, London, UK; Translational Health Sciences, School of Clinical Sciences, University of Bristol, Bristol, UK.
| |
Collapse
|
48
|
Rosada C, Bauer M, Golde S, Metz S, Roepke S, Otte C, Wolf OT, Buss C, Wingenfeld K. Association between childhood trauma and brain anatomy in women with post-traumatic stress disorder, women with borderline personality disorder, and healthy women. Eur J Psychotraumatol 2021; 12:1959706. [PMID: 34567441 PMCID: PMC8462923 DOI: 10.1080/20008198.2021.1959706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
BACKGROUND Childhood trauma (CT) is associated with altered brain anatomy. These neuroanatomical changes might be more pronounced in individuals with a psychiatric disorder. Post-traumatic stress disorder (PTSD) and borderline personality disorder (BPD) are more prevalent in individuals with a history of CT. OBJECTIVE In this study, we examined limbic and total brain volumes in healthy women with and without a history of CT and in females with PTSD or BPD and a history of CT to see whether neuroanatomical changes are a function of psychopathology or CT. METHOD In total, 128 women (N = 70 healthy controls without CT, N = 25 healthy controls with CT, N = 14 individuals with PTSD, and N = 19 individuals with BPD) were recruited. A T1-weighted anatomical MRI was acquired from all participants for Freesurfer-based assessment of total brain, hippocampus, and amygdala volumes. Severity of CT was assessed with a clinical interview and the Childhood Trauma Questionnaire. Group differences in hippocampal and amygdala volumes (adjusted for total brain volume) and total brain volume (adjusted for height) were characterized by analysis of covariance. RESULTS Volume of the total brain, hippocampus, and amygdala did not differ between the four groups (p > .05). CT severity correlated negatively with total brain volume across groups (r = -0.20; p = .029). CONCLUSIONS CT was associated with reduced brain volume but PTSD or BPD was not. The association between CT and reduced brain volume as a global measure of brain integrity suggests a common origin for vulnerability to psychiatric disorders later in life.
Collapse
Affiliation(s)
- Catarina Rosada
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Klinik Für Psychiatrie Und Psychotherapie, Campus Benjamin Franklin, Berlin, Germany
| | - Martin Bauer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität Zu Berlin, and Berlin Institute of Health, Institute of Medical Psychology, Berlin, Germany
| | - Sabrina Golde
- Clinical Psychology and Psychotherapy, Department of Education and Psychology, Freie Universität, Berlin, Germany
| | - Sophie Metz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Klinik Für Psychiatrie Und Psychotherapie, Campus Benjamin Franklin, Berlin, Germany
| | - Stefan Roepke
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Klinik Für Psychiatrie Und Psychotherapie, Campus Benjamin Franklin, Berlin, Germany
| | - Christian Otte
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Klinik Für Psychiatrie Und Psychotherapie, Campus Benjamin Franklin, Berlin, Germany
| | - Oliver T Wolf
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Claudia Buss
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität Zu Berlin, and Berlin Institute of Health, Institute of Medical Psychology, Berlin, Germany.,Department of Pediatrics, University of California, Irvine, CA, USA; Development, Health and Disease Research Program, University of California, Irvine, CA, USA
| | - Katja Wingenfeld
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Klinik Für Psychiatrie Und Psychotherapie, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
49
|
Abstract
Stress system dysfunction is a typical characteristic of acute depression and other mood disorders. The exact pattern of factors predisposing for stress-related mental disorders is yet to be unraveled. However, corticosteroid receptor function plays an important role for appropriate or dysfunctional neuroendocrine responses to stress exposure and hence in resilience or risk for the development and course of both, depression and anxiety disorders. Solid neuroscience data strongly support that both neuropeptides, corticotropin-releasing hormone (CRH) and vasopressin (AVP), are central in coordinating humoral and behavioral adaptation to stress. Other neuropeptides, including oxytocin, neuropeptide S, neuropeptide Y, and orexin, are also considered important contributors. Attempts to turn neuropeptide biology into treatments for stress-related disorders need to consider that neuropeptide receptors are specific drug targets for certain patient populations rather than universal targets for all patients, like biogenic amine systems. That is why most negative clinical trials testing neuropeptide receptor antagonists have been in fact failed trials by design, because no companion tests were used to identify which patients with depression are most likely to benefit from a specific neuropeptide receptor-targeting drug treatment. Therefore, the most important future research task is discovery and development of appropriate companion tests that will allow the successful transfer of the precious treasure of neuropeptide system-targeting drugs into clinics.
Collapse
Affiliation(s)
| | - Marcus Ising
- Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
50
|
Sjörs Dahlman A, Jonsdottir IH, Hansson C. The hypothalamo-pituitary-adrenal axis and the autonomic nervous system in burnout. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:83-94. [PMID: 34266613 DOI: 10.1016/b978-0-12-819973-2.00006-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Burnout constitutes a serious health concern in the modern working environment. It is a stress-related condition that has developed as a result of a prolonged psychosocial stress exposure causing a persistent mismatch between demands and resources. The main symptom is emotional exhaustion, but physical fatigue, diminished professional efficacy, cynicism, and cognitive impairments are also associated with this condition. Burnout has been used both as a psychologic term in occupational settings and as a clinical diagnosis in patient populations, and there is currently no universally accepted definition and diagnostic criteria of burnout. It has been hypothesized that the two main stress response systems, the autonomic nervous system (ANS) and the hypothalamus-pituitary-adrenal axis (HPA axis), are involved in the pathogenesis of burnout. A common hypothesis is that in the early stages of chronic stress, the HPA axis and sympathetic ANS activity tend to be higher, while it will decrease with a longer duration of chronic stress to ultimately reach a state of hypoactivity in clinical burnout. The current research in this field shows many contradictory results. Thus there is no compelling evidence of either ANS or HPA dysfunction in burnout. However, there is partial support for the hypothesis of HPA and sympathetic hyperactivity in early stages, and HPA hyporeactivity and low vagal activity in more severe burnout cases, but high-quality studies investigating the causal links are still lacking.
Collapse
Affiliation(s)
- Anna Sjörs Dahlman
- Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; Human Factors Department, Swedish National Road and Transport Research Institute, Gothenburg, Sweden.
| | - Ingibjörg H Jonsdottir
- Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Caroline Hansson
- Institute of Stress Medicine, Region Västra Götaland, Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|