1
|
Ghaffarzadegan R, Akhondzadeh S, Nikasa Z, Hajizamani S, Mehrabanifar S, Cheraghi I, Vaseghi S. New Insights into Contradictory Changes in Brain-Derived Neurotrophic Factor (BDNF) in Rodent Models of Posttraumatic Stress Disorder (PTSD). Neurochem Res 2024; 49:3226-3243. [PMID: 39283581 DOI: 10.1007/s11064-024-04242-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/31/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a neuropsychiatric disorder that may develop after experiencing traumatic events. Preclinical studies use various methods to induce PTSD-like models such as fear-conditioning, single-prolonged stress (SPS), restraint stress, and social defeat. Brain-derived neurotrophic factor (BDNF) is a crucial neurotrophin in mood regulation. Evidence shows BDNF changes in different neuropsychiatric disorders particularly PTSD. This review examined BDNF alterations in preclinical rodent models of PTSD where we demonstrated a wide range of paradoxical changes in BDNF. We found that the fear-conditioning model produced the most inconsistent alterations in BDNF, and suggest that conclusions drawn from these changes be approached with caution. We suggest that BDNF maladaptive changes in social defeat and restraint stress models may be related to the duration of stress, while the SPS model appears to have more consistent results. Ultimately, we propose that evaluating BDNF alterations in the process of treating PTSD symptoms may not be a reliable factor.
Collapse
Affiliation(s)
- Reza Ghaffarzadegan
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, P.O. Box: 1419815477, Karaj, Iran
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Nikasa
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Shadi Hajizamani
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Iman Cheraghi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Salar Vaseghi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, P.O. Box: 1419815477, Karaj, Iran.
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
| |
Collapse
|
2
|
Tofani GSS, Leigh SJ, Gheorghe CE, Bastiaanssen TFS, Wilmes L, Sen P, Clarke G, Cryan JF. Gut microbiota regulates stress responsivity via the circadian system. Cell Metab 2024:S1550-4131(24)00399-1. [PMID: 39504963 DOI: 10.1016/j.cmet.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 04/12/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024]
Abstract
Stress and circadian systems are interconnected through the hypothalamic-pituitary-adrenal (HPA) axis to maintain responses to external stimuli. Yet, the mechanisms of how such signals are orchestrated remain unknown. Here, we uncover the gut microbiota as a regulator of HPA-axis rhythmicity. Microbial depletion disturbs the brain transcriptome and metabolome in stress-responding pathways in the hippocampus and amygdala across the day. This is coupled with a dysregulation of the circadian pacemaker in the brain that results in perturbed glucocorticoid rhythmicity. The resulting hyper-activation of the HPA axis at the sleep/wake transition drives time-of-day-specific impairments of the stress response and stress-sensitive behaviors. Finally, microbiota transplantation confirmed that diurnal oscillations of gut microbes underlie altered glucocorticoid secretion and that L. reuteri is a candidate strain for such effects. Our data offer compelling evidence that the microbiota regulates stress responsiveness in a circadian manner and is necessary to respond adaptively to stressors throughout the day.
Collapse
Affiliation(s)
- Gabriel S S Tofani
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | - Sarah-Jane Leigh
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry & Neurobehavioral Sciences, University College Cork, Cork, Ireland
| | - Cassandra E Gheorghe
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry & Neurobehavioral Sciences, University College Cork, Cork, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | - Lars Wilmes
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry & Neurobehavioral Sciences, University College Cork, Cork, Ireland
| | - Paromita Sen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry & Neurobehavioral Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
3
|
Brivio P, Gallo MT, Audano M, Galassi G, Gruca P, Lason M, Litwa E, Fumagalli F, Papp M, Mitro N, Calabrese F. Exposure to an acute stress impaired the metabolic plasticity of resilient rats by enhancing fatty acid β-oxidation in the ventral hippocampus. Transl Psychiatry 2024; 14:366. [PMID: 39256372 PMCID: PMC11387825 DOI: 10.1038/s41398-024-03080-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
The concept of resilience has changed over time and nowadays it refers to the positive adaptation to life adversities, rather than to the absence of a pathological response normally occurring in susceptible people. Based on our previous data showing that the exposure to the chronic mild stress (CMS) paradigm differently affected bioenergetics in the ventral hippocampus of vulnerable and resilient animals, here we investigated whether resilience is a stable trait or if the energetic strategy set in motion to sustain resilience unveils a vulnerability feature in a more dynamic situation. To this aim, vulnerable and resilient rats after 6 weeks of CMS were subjected to a further acute, unfamiliar restraint stress (ARS) and metabolomic studies were conducted in the ventral hippocampus. We observed that exposure to a single novel challenge negatively affects the fuel utilization of resilient animals. Indeed, while they increase glycolysis to sustain the non-hedonic phenotype when exposed to CMS, they shift to fatty acid β-oxidation after ARS, as vulnerable animals following CMS, suggesting that the energy strategy that guarantees resilience is fragile and can be negatively modified by a different environmental condition. These results suggest that strengthening resilience to foster individuals to bounce back from stressful life events may represent a strategy to decrease vulnerability or prevent the risk of relapsing to a pathological state.
Collapse
Affiliation(s)
- Paola Brivio
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Maria Teresa Gallo
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Gaia Galassi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Piotr Gruca
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Magdalena Lason
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewa Litwa
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
4
|
Lightman SL, Conway-Campbell BL. Circadian and ultradian rhythms: Clinical implications. J Intern Med 2024; 296:121-138. [PMID: 38825772 DOI: 10.1111/joim.13795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The hypothalamic-pituitary-adrenal axis is an extremely dynamic system with a combination of both circadian and ultradian oscillations. This state of 'continuous dynamic equilibration' provides a platform that is able to anticipate events, is sensitive in its response to stressors, remains robust during perturbations of both the internal and external environments and shows plasticity to adapt to a changed environment. In this review, we describe these oscillations of glucocorticoid (GC) hormones and why they are so important for GC-dependent gene activation in the brain and liver, and their consequent effects on the regulation of synaptic and memory function as well as appetite control and metabolic regulation. Abnormalities of mood, appetite and metabolic regulation are well-known consequences of GC therapy, and we suggest that the pattern of GC treatment and hormone replacement should be a much higher priority for endocrinologists and the pharmaceutical industry. One of the major impediments to our research on the importance of these cortisol rhythms in our patients has been our inability to measure repeated levels of hormones across the day in patients in their home or work surroundings. We describe how new wearable methodologies now allow the measurement of 24-h cortisol profiles - including during sleep - and will enable us to define physiological normality and allow us both to develop better diagnostic tests and inform, at an individual patient level, how to improve replacement therapy.
Collapse
Affiliation(s)
- Stafford L Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Becky L Conway-Campbell
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
5
|
Wei B, Shi Y, Yu X, Cai Y, Zhao Y, Song Y, Zhao Z, Huo M, Li L, Gao Q, Yu D, Wang B, Sun M. GR/P300 Regulates MKP1 Signaling Pathway and Mediates Depression-like Behavior in Prenatally Stressed Offspring. Mol Neurobiol 2024:10.1007/s12035-024-04244-y. [PMID: 38769227 DOI: 10.1007/s12035-024-04244-y] [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: 10/13/2023] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Accumulating evidence suggests that prenatal stress (PNS) increases offspring susceptibility to depression, but the underlying mechanisms remain unclear. We constructed a mouse model of prenatal stress by spatially restraining pregnant mice from 09:00-11:00 daily on Days 5-20 of gestation. In this study, western blot analysis, quantitative real-time PCR (qRT‒PCR), immunofluorescence, immunoprecipitation, chromatin immunoprecipitation (ChIP), and mifepristone rescue assays were used to investigate alterations in the GR/P300-MKP1 and downstream ERK/CREB/TRKB pathways in the brains of prenatally stressed offspring to determine the pathogenesis of the reduced neurogenesis and depression-like behaviors in offspring induced by PNS. We found that prenatal stress leads to reduced hippocampal neurogenesis and depression-like behavior in offspring. Prenatal stress causes high levels of glucocorticoids to enter the fetus and activate the hypothalamic‒pituitary‒adrenal (HPA) axis, resulting in decreased hippocampal glucocorticoid receptor (GR) levels in offspring. Furthermore, the nuclear translocation of GR and P300 (an acetylation modifying enzyme) complex in the hippocampus of PNS offspring increased significantly. This GR/P300 complex upregulates MKP1, which is a negative regulator of the ERK/CREB/TRKB signaling pathway associated with depression. Interestingly, treatment with a GR antagonist (mifepristone, RU486) increased hippocampal GR levels and decreased MKP1 expression, thereby ameliorating abnormal neurogenesis and depression-like behavior in PNS offspring. In conclusion, our study suggested that the regulation of the MKP1 signaling pathway by GR/P300 is involved in depression-like behavior in prenatal stress-exposed offspring and provides new insights and ideas for the fetal hypothesis of mental health.
Collapse
Affiliation(s)
- Bin Wei
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250000, Shandong, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yajun Shi
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Xi Yu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yongle Cai
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yan Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yueyang Song
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Zejun Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Ming Huo
- Reproductive Medicine Center, The First Hospital of Lanzhou University, LanzhouGansu, 730000, China
| | - Lingjun Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Qinqin Gao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Dongyi Yu
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250000, Shandong, China
| | - Bin Wang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China.
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China.
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory for Complex Severe and Rare Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, 100005, China.
| |
Collapse
|
6
|
De Gasperi R, Gama Sosa MA, Perez Garcia G, Perez GM, Pryor D, Morrison CLA, Lind R, Abutarboush R, Kawoos U, Statz JK, Patterson J, Hof PR, Zhu CW, Ahlers ST, Cook DG, Elder GA. Metabotropic Glutamate Receptor 2 Expression Is Chronically Elevated in Male Rats With Post-Traumatic Stress Disorder Related Behavioral Traits Following Repetitive Low-Level Blast Exposure. J Neurotrauma 2024; 41:714-733. [PMID: 37917117 PMCID: PMC10902502 DOI: 10.1089/neu.2023.0252] [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] [Indexed: 11/03/2023] Open
Abstract
Many military veterans who experienced blast-related traumatic brain injuries in the conflicts in Iraq and Afghanistan currently suffer from chronic cognitive and mental health problems that include depression and post-traumatic stress disorder (PTSD). Male rats exposed to repetitive low-level blast develop cognitive and PTSD-related behavioral traits that are present for more than 1 year after exposure. We previously reported that a group II metabotropic receptor (mGluR2/3) antagonist reversed blast-induced behavioral traits. In this report, we explored mGluR2/3 expression following blast exposure in male rats. Western blotting revealed that mGluR2 protein (but not mGluR3) was increased in all brain regions studied (anterior cortex, hippocampus, and amygdala) at 43 or 52 weeks after blast exposure but not at 2 weeks or 6 weeks. mGluR2 RNA was elevated at 52 weeks while mGluR3 was not. Immunohistochemical staining revealed no changes in the principally presynaptic localization of mGluR2 by blast exposure. Administering the mGluR2/3 antagonist LY341495 after behavioral traits had emerged rapidly reversed blast-induced effects on novel object recognition and cued fear responses 10 months following blast exposure. These studies support alterations in mGluR2 receptors as a key pathophysiological event following blast exposure and provide further support for group II metabotropic receptors as therapeutic targets in the neurobehavioral effects that follow blast injury.
Collapse
Affiliation(s)
- Rita De Gasperi
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Georgina Perez Garcia
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gissel M. Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Dylan Pryor
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Chenel L-A. Morrison
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Northeast Regional Alliance Health Careers Opportunity Program, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rachel Lind
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Rania Abutarboush
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Jonathan K. Statz
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland, USA
| | - Jacob Patterson
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
| | - Patrick R. Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carolyn W. Zhu
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Naval Medical Research Command, Silver Spring, Maryland, USA
| | - David G. Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Medicine, Pharmacology, and Psychiatry, University of Washington, Seattle, Washington, USA
- Department of Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Gregory A. Elder
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| |
Collapse
|
7
|
Ell MA, Schiele MA, Iovino N, Domschke K. Epigenetics of Fear, Anxiety and Stress - Focus on Histone Modifications. Curr Neuropharmacol 2024; 22:843-865. [PMID: 36946487 PMCID: PMC10845084 DOI: 10.2174/1570159x21666230322154158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 03/23/2023] Open
Abstract
Fear-, anxiety- and stress-related disorders are among the most frequent mental disorders. Given substantial rates of insufficient treatment response and often a chronic course, a better understanding of the pathomechanisms of fear-, anxiety- and stress-related disorders is urgently warranted. Epigenetic mechanisms such as histone modifications - positioned at the interface between the biological and the environmental level in the complex pathogenesis of mental disorders - might be highly informative in this context. The current state of knowledge on histone modifications, chromatin-related pharmacology and animal models modified for genes involved in the histone-related epigenetic machinery will be reviewed with respect to fear-, anxiety- and stress-related states. Relevant studies, published until 30th June 2022, were identified using a multi-step systematic literature search of the Pub- Med and Web of Science databases. Animal studies point towards histone modifications (e.g., H3K4me3, H3K9me1/2/3, H3K27me2/3, H3K9ac, H3K14ac and H4K5ac) to be dynamically and mostly brain region-, task- and time-dependently altered on a genome-wide level or gene-specifically (e.g., Bdnf) in models of fear conditioning, retrieval and extinction, acute and (sub-)chronic stress. Singular and underpowered studies on histone modifications in human fear-, anxiety- or stress-related phenotypes are currently restricted to the phenotype of PTSD. Provided consistent validation in human phenotypes, epigenetic biomarkers might ultimately inform indicated preventive interventions as well as personalized treatment approaches, and could inspire future innovative pharmacological treatment options targeting the epigenetic machinery improving treatment response in fear-, anxiety- and stressrelated disorders.
Collapse
Affiliation(s)
- Marco A. Ell
- Department of Psychiatry and Psychotherapy, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Miriam A. Schiele
- Department of Psychiatry and Psychotherapy, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Nicola Iovino
- Department of Chromation Regulation, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
8
|
Yang N, Wang Y, Luo X, Zhan G. Chronic restraint stress induces abnormal behaviors in pain sensitivity and cognitive function in mice: the role of Keap1/Nrf2 pathway. Stress 2024; 27:2316050. [PMID: 38377152 DOI: 10.1080/10253890.2024.2316050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/03/2024] [Indexed: 02/22/2024] Open
Abstract
Stress is a series of physical and psychological responses to external and internal environmental stimuli. Growing studies have demonstrated the detrimental impacts of acute restraint stress (ARS) and chronic restraint stress (CRS) on animal behavior. However, the related pathogenesis and therapeutic mechanisms remain unclear. Hence, the present study aimed to examine whether unfolded protein response (UPR) and Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2 related factor 2 (Nrf2) pathway are associated with ARS- and CRS- induced abnormal behaviors of pain sensitivity and cognitive function. We here used four behavioral tests to evaluate pain sensitivity and cognitive function in ARS and CRS mice. CRS markedly decreased Paw Withdrawal Mechanical Threshold (PWMT) and Tail-flick Latency (TFL) scores, whereas ARS altered TFL but had no effect on PWMT scores. Additionally, CRS, but not ARS, significantly changed behaviors in nest building behavior and MWMT. Intriguingly, the expression of Keap1 and Nrf2 protein were decreased in the spinal cord and hippocampus in CRS mice, but not in ARS mice. Moreover, neither the ARS nor the CRS groups significantly differed from the control group in terms of endoplasmic reticulum stress (ERS). Taken together, this study demonstrated that CRS could induce abnormal pain sensitivity and cognitive function probably via Keap1/Nrf2 pathway in spinal cord and hippocampus. It is therefore likely that effective intervention of Keap1/Nrf2 pathway may contribute to preventing and treating hyperalgesia and cognitive dysfunction in CRS.
Collapse
Affiliation(s)
- Ning Yang
- Department of Anesthesiology, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Wang
- Department of Anesthesiology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Borges Dos Santos JR, Rae M, Teixeira SA, Muscará MN, Szumlinski KK, Camarini R. The effect of MK-801 on stress-ethanol cross-sensitization is dissociable from its effects on nNOS activity. Alcohol 2023; 112:31-39. [PMID: 37479092 DOI: 10.1016/j.alcohol.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/23/2023]
Abstract
Locomotor behavioral sensitization represents an animal model for understanding neuroadaptive processes related to repeated drug exposure. Repeated stress can elicit a cross-sensitization to the stimulant response of ethanol, which involves neuronal nitric oxide synthase (nNOS). Activation of N-methyl d-aspartate (NMDA) glutamate receptors triggers nNOS and the synthesis of nitric oxide (NO). In this study, we investigated the effects of blocking NMDA receptors using the NMDA receptor antagonist MK-801 on the cross-sensitization between restraint stress and ethanol. We also evaluated the nNOS activity in the prefrontal cortex (PFC) and hippocampus. Mice were pretreated with saline or MK-801 30 min before an injection of saline or stress exposure for 14 days. On the following day, they were challenged with either saline or 1.8 g/kg ethanol. Swiss male mice pretreated with 0.25 mg/kg MK-801 exhibited a sensitized response to ethanol. Moreover, MK-801 potentiated the cross-sensitization between stress and ethanol. However, MK-801 prevented the enhanced nNOS activity in stress-exposed groups (challenged with saline or ethanol) in the PFC; the antagonist also prevented the ethanol-induced increase in nNOS activity and reduced this enzyme activity in mice exposed to stress in the hippocampus. These data indicate that systemic treatment with the NMDA antagonist potentiated, rather than blocked, ethanol-induced behavioral sensitization and that this effect is dissociable from the capacity of NMDA antagonists to reduce ethanol/stress-induced NOS stimulation in the PFC and hippocampus.
Collapse
Affiliation(s)
- Jaqueline Rocha Borges Dos Santos
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, SP, Brazil; Department of Pharmaceutical Sciences, Institute of Biological and Health Sciences, Universidade Federal Rural Do Rio de Janeiro, RJ, Brazil
| | - Mariana Rae
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, SP, Brazil
| | | | - Marcelo Nicolás Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, SP, Brazil
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, Department of Molecular, Cellular and Developmental Biology and the Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, United States
| | - Rosana Camarini
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, SP, Brazil.
| |
Collapse
|
10
|
McNearney TA, Westlund KN. Pluripotential GluN1 (NMDA NR1): Functional Significance in Cellular Nuclei in Pain/Nociception. Int J Mol Sci 2023; 24:13196. [PMID: 37686003 PMCID: PMC10488196 DOI: 10.3390/ijms241713196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The N-methyl-D-aspartate (NMDA) glutamate receptors function as plasma membrane ionic channels and take part in very tightly controlled cellular processes activating neurogenic and inflammatory pathways. In particular, the NR1 subunit (new terminology: GluN1) is required for many neuronal and non-neuronal cell functions, including plasticity, survival, and differentiation. Physiologic levels of glutamate agonists and NMDA receptor activation are required for normal neuronal functions such as neuronal development, learning, and memory. When glutamate receptor agonists are present in excess, binding to NMDA receptors produces neuronal/CNS/PNS long-term potentiation, conditions of acute pain, ongoing severe intractable pain, and potential excitotoxicity and pathology. The GluNR1 subunit (116 kD) is necessary as the anchor component directing ion channel heterodimer formation, cellular trafficking, and the nuclear localization that directs functionally specific heterodimer formation, cellular trafficking, and nuclear functions. Emerging studies report the relevance of GluN1 subunit composition and specifically that nuclear GluN1 has major physiologic potential in tissue and/or subnuclear functioning assignments. The shift of the GluN1 subunit from a surface cell membrane to nuclear localization assigns the GluN1 promoter immediate early gene behavior with access to nuclear and potentially nucleolar functions. The present narrative review addresses the nuclear translocation of GluN1, focusing particularly on examples of the role of GluN1 in nociceptive processes.
Collapse
Affiliation(s)
- Terry A. McNearney
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch Galveston, Galveston, TX 77555-1043, USA;
- Department of Internal Medicine, University of Texas Medical Branch Galveston, Galveston, TX 77555-1043, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, Galveston, TX 77555-1043, USA
| | - Karin N. Westlund
- Department of Anesthesiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
- Biomedical Laboratory Research & Development (121F), New Mexico VA Health Care System, Albuquerque, NM 87108-5153, USA
| |
Collapse
|
11
|
Asraf K, Zaidan H, Natoor B, Gaisler-Salomon I. Synergistic, long-term effects of glutamate dehydrogenase 1 deficiency and mild stress on cognitive function and mPFC gene and miRNA expression. Transl Psychiatry 2023; 13:248. [PMID: 37419882 DOI: 10.1038/s41398-023-02534-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
Glutamate abnormalities in the medial prefrontal cortex (mPFC) are associated with cognitive deficits. We previously showed that homozygous deletion of CNS glutamate dehydrogenase 1 (Glud1), a metabolic enzyme critical for glutamate metabolism, leads to schizophrenia-like behavioral abnormalities and increased mPFC glutamate; mice heterozygous for CNS Glud1 deletion (C-Glud1+/- mice) showed no cognitive or molecular abnormalities. Here, we examined the protracted behavioral and molecular effects of mild injection stress on C-Glud1+/- mice. We found spatial and reversal learning deficits, as well as large-scale mPFC transcriptional changes in pathways associated with glutamate and GABA signaling, in stress-exposed C-Glud1+/- mice, but not in their stress-naïve or C-Glud1+/+ littermates. These effects were observed several weeks following stress exposure, and the expression levels of specific glutamatergic and GABAergic genes differentiated between high and low reversal learning performance. An increase in miR203-5p expression immediately following stress may provide a translational regulatory mechanism to account for the delayed effect of stress exposure on cognitive function. Our findings show that chronic glutamate abnormalities interact with acute stress to induce cognitive deficits, and resonate with gene x environment theories of schizophrenia. Stress-exposed C-Glud1+/- mice may model a schizophrenia high-risk population, which is uniquely sensitive to stress-related 'trigger' events.
Collapse
Affiliation(s)
- Kfir Asraf
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Hiba Zaidan
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Baylasan Natoor
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel
| | - Inna Gaisler-Salomon
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa, 3498838, Israel.
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
12
|
Torrisi SA, Rizzo S, Laudani S, Ieraci A, Drago F, Leggio GM. Acute stress alters recognition memory and AMPA/NMDA receptor subunits in a sex-dependent manner. Neurobiol Stress 2023; 25:100545. [PMID: 37293561 PMCID: PMC10244889 DOI: 10.1016/j.ynstr.2023.100545] [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: 03/06/2023] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Several studies have consistently reported a detrimental effect of chronic stress on recognition memory. However, the effects of acute stress on this cognitive ability have been poorly investigated. Moreover, despite well-documented sex differences in recognition memory observed in clinical studies, most of the preclinical studies in this field of research have been carried out by using solely male rodents. Here we tested the hypothesis that acute stress could affect the consolidation of different types of recognition memory in a sex-dependent manner. For this purpose, male and female C57BL6/J mice were exposed to 2-h of restrain stress immediately after the training session of both the novel object recognition (NOR) test and novel object location (NOL) tasks. Acute restraint stress did not affect memory performance of male and female mice, after a 4-h delay between the training session and the test phase of both tasks. By contrast, acute restraint stress altered memory performance in a sex-dependent manner, after a 24-h delay. While stressed mice of both sexes were impaired in the NOL test, only male stressed mice were impaired in the NOR test. Because ionotropic glutamate receptors-mediated neurotransmission is essential for shaping recognition memory, we further tested the hypothesis that post training acute stress could induce sex-dependent transcriptional changes of ionotropic glutamate receptor subunits in the dorsal hippocampus. We uncovered that acute stress induced sex-, time- and type of memory-dependent transcriptional changes of N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits. These findings suggest that the effect of acute stress on recognition memory can be strongly biased by multiple factors including sex. These findings also indicate that the same stress-induced memory impairment observed in both sexes can be triggered by different sex-dependent molecular mechanisms. At the therapeutic level, this should not be overlooked in the context of personalized and targeted treatments.
Collapse
Affiliation(s)
- Sebastiano A. Torrisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Silvia Rizzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Samuele Laudani
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Alessandro Ieraci
- Department of Theoretical and Applied Sciences, eCampus University, 22060, Novedrate, CO, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Gian Marco Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
13
|
Martini P, Mingardi J, Carini G, Mattevi S, Ndoj E, La Via L, Magri C, Gennarelli M, Russo I, Popoli M, Musazzi L, Barbon A. Transcriptional Profiling of Rat Prefrontal Cortex after Acute Inescapable Footshock Stress. Genes (Basel) 2023; 14:genes14030740. [PMID: 36981011 PMCID: PMC10048409 DOI: 10.3390/genes14030740] [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: 12/30/2022] [Revised: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Stress is a primary risk factor for psychiatric disorders such as Major Depressive Disorder (MDD) and Post Traumatic Stress Disorder (PTSD). The response to stress involves the regulation of transcriptional programs, which is supposed to play a role in coping with stress. To evaluate transcriptional processes implemented after exposure to unavoidable traumatic stress, we applied microarray expression analysis to the PFC of rats exposed to acute footshock (FS) stress that were sacrificed immediately after the 40 min session or 2 h or 24 h after. While no substantial changes were observed at the single gene level immediately after the stress session, gene set enrichment analysis showed alterations in neuronal pathways associated with glia development, glia-neuron networking, and synaptic function. Furthermore, we found alterations in the expression of gene sets regulated by specific transcription factors that could represent master regulators of the acute stress response. Of note, these pathways and transcriptional programs are activated during the early stress response (immediately after FS) and are already turned off after 2 h-while at 24 h, the transcriptional profile is largely unaffected. Overall, our analysis provided a transcriptional landscape of the early changes triggered by acute unavoidable FS stress in the PFC of rats, suggesting that the transcriptional wave is fast and mild, but probably enough to activate a cellular response to acute stress.
Collapse
Affiliation(s)
- Paolo Martini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Jessica Mingardi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Giulia Carini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25123 Brescia, Italy
| | - Stefania Mattevi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Elona Ndoj
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Luca La Via
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Chiara Magri
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25123 Brescia, Italy
| | - Isabella Russo
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25123 Brescia, Italy
| | - Maurizio Popoli
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Laura Musazzi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Alessandro Barbon
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
14
|
Tseilikman VE, Tseilikman OB, Pashkov AA, Ivleva IS, Karpenko MN, Shatilov VA, Zhukov MS, Fedotova JO, Kondashevskaya MV, Downey HF, Manukhina EB. Mechanisms of Susceptibility and Resilience to PTSD: Role of Dopamine Metabolism and BDNF Expression in the Hippocampus. Int J Mol Sci 2022; 23:ijms232314575. [PMID: 36498900 PMCID: PMC9737079 DOI: 10.3390/ijms232314575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Susceptibility and resilience to post-traumatic stress disorder (PTSD) are recognized, but their mechanisms are not understood. Here, the hexobarbital sleep test (HST) was used to elucidate mechanisms of PTSD resilience or susceptibility. A HST was performed in rats 30 days prior to further experimentation. Based on the HST, the rats were divided into groups: (1) fast metabolizers (FM; sleep duration < 15 min); (2) slow metabolizers (SM; sleep duration ≥ 15 min). Then the SM and FM groups were subdivided into stressed (10 days predator scent, 15 days rest) and unstressed subgroups. Among stressed animals, only SMs developed experimental PTSD, and had higher plasma corticosterone (CORT) than stressed FMs. Thus, resilience or susceptibility to PTSD was consistent with changes in glucocorticoid metabolism. Stressed SMs had a pronounced decrease in hippocampal dopamine associated with increased expressions of catecholamine-O-methyl-transferase and DA transporter. In stressed SMs, a decrease in monoaminoxidase (MAO) A was associated with increased expressions of hippocampal MAO-A and MAO-B. BDNF gene expression was increased in stressed FMs and decreased in stressed SMs. These results demonstrate relationships between the microsomal oxidation phenotype, CORT concentration, and anxiety, and they help further the understanding of the role of the liver−brain axis during PTSD.
Collapse
Affiliation(s)
- Vadim E. Tseilikman
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
| | - Olga B. Tseilikman
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
- Department of Basic Medicine, Chelyabinsk State University, 454001 Chelyabinsk, Russia
| | - Anton A. Pashkov
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
- Federal Neurosurgical Center, 630048 Novosibirsk, Russia
| | - Irina S. Ivleva
- Pavlov Department of Physiology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia
| | - Marina N. Karpenko
- Pavlov Department of Physiology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia
| | | | - Maxim S. Zhukov
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
| | - Julia O. Fedotova
- Laboratory of Neuroendocrinology, Pavlov Institute of Physiology, 199034 Saint Petersburg, Russia
| | - Marina V. Kondashevskaya
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 117418 Moscow, Russia
| | - H. Fred Downey
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Correspondence:
| | - Eugenia B. Manukhina
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Laboratory for Regulatory Mechanisms of Stress and Adaptation, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| |
Collapse
|
15
|
Helman TJ, Headrick JP, Vider J, Peart JN, Stapelberg NJC. Sex-specific behavioral, neurobiological, and cardiovascular responses to chronic social stress in mice. J Neurosci Res 2022; 100:2004-2027. [PMID: 36059192 DOI: 10.1002/jnr.25115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023]
Abstract
Psychosocial stress promotes and links mood and cardiovascular disorders in a sex-specific manner. However, findings in animal models are equivocal, in some cases opposing human dimorphisms. We examined central nervous system (CNS), behavioral, endocrine, cardiac, and hepatic outcomes in male or female C57Bl/6 mice subjected to chronic social stress (56 days of social isolation, with intermittent social confrontation encounters twice daily throughout the final 20 days). Females exhibited distinct physiological and behavioral changes, including relative weight loss, and increases in coronary resistance, hepatic inflammation, and thigmotaxic behavior in the open field. Males evidence reductions in coronary resistance and cardiac ischemic tolerance, with increased circulating and hippocampal monoamine levels and emerging anhedonia. Shared CNS gene responses include reduced hippocampal Maoa and increased Htr1b expression, while unique responses include repression of hypothalamic Ntrk1 and upregulation of cortical Nrf2 and Htr1b in females; and repression of hippocampal Drd1 and hypothalamic Gabra1 and Oprm in males. Declining cardiac stress resistance in males was associated with repression of cardiac leptin levels and metabolic, mitochondrial biogenesis, and anti-inflammatory gene expression. These integrated data reveal distinct biological responses to social stress in males and females, and collectively evidence greater biological disruption or allostatic load in females (consistent with propensities to stress-related mood and cardiovascular disorders in humans). Distinct stress biology, and molecular to organ responses, emphasize the importance of sex-specific mechanisms and potential approaches to stress-dependent disease.
Collapse
Affiliation(s)
- Tessa J Helman
- School of Pharmacy and Medical Sciences, Griffith University, Southport, Queensland, Australia
| | - John P Headrick
- School of Pharmacy and Medical Sciences, Griffith University, Southport, Queensland, Australia
| | - Jelena Vider
- School of Pharmacy and Medical Sciences, Griffith University, Southport, Queensland, Australia
| | - Jason N Peart
- School of Pharmacy and Medical Sciences, Griffith University, Southport, Queensland, Australia
| | - Nicolas J C Stapelberg
- Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia.,Gold Coast Hospital and Health Service, Southport, Queensland, Australia
| |
Collapse
|
16
|
Tyler RE, Besheer J, Joffe ME. Advances in translating mGlu 2 and mGlu 3 receptor selective allosteric modulators as breakthrough treatments for affective disorders and alcohol use disorder. Pharmacol Biochem Behav 2022; 219:173450. [PMID: 35988792 PMCID: PMC10405528 DOI: 10.1016/j.pbb.2022.173450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/26/2022] [Accepted: 08/13/2022] [Indexed: 11/16/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are promising targets for the treatment of affective disorders and alcohol use disorder (AUD). Nonspecific ligands for Group II (mGlu2 and mGlu3) mGlu receptors have demonstrated consistent therapeutic potential for affective disorders in preclinical models. Disentangling the specific roles of mGlu2 versus mGlu3 receptors in these effects has persisted as a major challenge, in part due to pharmacological limitations. However, the recent development of highly specific allosteric modulators for both mGlu2 and mGlu3 receptors have enabled straightforward and rigorous investigations into the specific function of each receptor. Here, we review recent experiments using these compounds that have demonstrated both similar and distinct receptor functions in behavioral, molecular, and electrophysiological measures associated with basal function and preclinical models of affective disorders. Studies using these selective drugs have demonstrated that mGlu2 is the predominant receptor subclass involved in presynaptic neurotransmitter release in prefrontal cortex. By contrast, the activation of postsynaptic mGlu3 receptors induces a cascade of cellular changes that results in AMPA receptor internalization, producing long-term depression and diminishing excitatory drive. Acute stress decreases the mGlu3 receptor function and dynamically alters transcript expression for both mGlu2 (Grm2) and mGlu3 (Grm3) receptors in brain areas involved in reward and stress. Accordingly, both mGlu2 and mGlu3 negative allosteric modulators show acute antidepressant-like effects and potential prophylactic effects against acute and traumatic stressors. The wide array of effects displayed by these new allosteric modulators of mGlu2 and mGlu3 receptors suggest that these drugs may act through improving endophenotypes of symptoms observed across several neuropsychiatric disorders. Therefore, recently developed allosteric modulators selective for mGlu2 or mGlu3 receptors show promise as potential therapeutics for affective disorders and AUD.
Collapse
Affiliation(s)
- Ryan E Tyler
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Neuroscience Curriculum, University of North Carolina at Chapel Hill, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, USA
| | - Joyce Besheer
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Neuroscience Curriculum, University of North Carolina at Chapel Hill, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, USA
| | - Max E Joffe
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219, USA; Translational Neuroscience Program, University of Pittsburgh, USA.
| |
Collapse
|
17
|
Brivio P, Gallo MT, Gruca P, Lason M, Litwa E, Fumagalli F, Papp M, Calabrese F. Resilience to chronic mild stress-induced anhedonia preserves the ability of the ventral hippocampus to respond to an acute challenge. Eur Arch Psychiatry Clin Neurosci 2022:10.1007/s00406-022-01470-0. [PMID: 36018382 PMCID: PMC10359391 DOI: 10.1007/s00406-022-01470-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 08/02/2022] [Indexed: 11/03/2022]
Abstract
Stress is a major precipitating factor for psychiatric disorders and its effects may depend on its duration and intensity. Of note, there are differences in individual susceptibility to stress, with some subjects displaying vulnerability and others showing resistance. Furthermore, the ability to react to stressful-life events can alter the response to a subsequent new stressor. Hence, we investigated whether the vulnerability and resilience to the chronic mild stress (CMS) paradigm, in terms of the hedonic phenotype, are paralleled by a different response when facing a novel acute challenge. Specifically, rats submitted to CMS were stratified based on their sucrose intake into vulnerable (anhedonic rats showing reduce intake of sucrose) and resilient (rats not showing the anhedonic-like behavior) subgroups and then further exposed to an acute restraint stress (ARS). Then, neuronal activation was investigated by measuring the gene expression of early immediate (IEG) genes such as Arc and Cfos and early response (ERG) genes, such as Gadd45β, Sgk1, Dusp1, and Nr4a1, in brain regions that play a crucial role in the stress response. We found that resilient rats preserve the ability to increase ERG expression following the ARS selectively in the ventral hippocampus. Conversely, such ability is lost in vulnerable rats. Interestingly, the recovery from the anhedonic phenotype observed in vulnerable rats after 3 weeks of rest from the CMS procedure also parallels the restoration of the ability to adequately respond to the challenge. In conclusion, these findings support the role of the ventral subregion of the hippocampus in the management of both chronic and acute stress response and point to this brain subregion as a critical target for a potential therapeutic strategy aimed at promoting stress resilience.
Collapse
Affiliation(s)
- Paola Brivio
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Maria Teresa Gallo
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Piotr Gruca
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Magdalena Lason
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewa Litwa
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
18
|
Tseilikman V, Akulov A, Shevelev O, Khotskina A, Kontsevaya G, Moshkin M, Fedotova J, Pashkov A, Tseilikman O, Agletdinov E, Tseilikman D, Kondashevskaya M, Zavjalov E. Paradoxical Anxiety Level Reduction in Animal Chronic Stress: A Unique Role of Hippocampus Neurobiology. Int J Mol Sci 2022; 23:ijms23169151. [PMID: 36012411 PMCID: PMC9409467 DOI: 10.3390/ijms23169151] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
A paradoxical reduction in anxiety levels in chronic predator stress paradigm (PS) in Sprague–Dawley rats has recently been shown in previous works. In this paper, we studied the possible neurobiological mechanism of this phenomenon. We segregated PS-exposed Sprague–Dawley rats into the high- and low-anxiety phenotypes. The long-lasting effects of PS on corticosterone levels, blood flow speed in the carotid arteries, diffusion coefficient, and 1H nuclear magnetic resonance spectra in the hippocampus were compared in the high-anxiety and low-anxiety rats. In addition, we evaluated the gene BDNF expression in the hippocampus which is considered to be a main factor of neuroplasticity. We demonstrated that in low-anxiety rats, the corticosterone level was decreased and carotid blood flow speed was increased. Moreover, in the hippocampus of low-anxiety rats compared to the control group and high-anxiety rats, the following changes were observed: (a) a decrease in N-acetyl aspartate levels with a simultaneous increase in phosphoryl ethanol amine levels; (b) an increase in lipid peroxidation levels; (c) a decrease in apparent diffusion coefficient value; (d) an increase in BDNF gene expression. Based on these findings, we proposed that stress-induced anxiety reduction is associated with the elevation of BDNF gene expression directly. Low corticosterone levels and a rise in carotid blood flow speed might facilitate BDNF gene expression. Meanwhile, the decrease in apparent diffusion coefficient value and decrease in N-acetyl aspartate levels, as well as an increase in the lipid peroxidation levels, in the hippocampus possibly reflected destructive changes in the hippocampus. We suggested that in Sprague–Dawley rats, these morphological alterations might be considered as an impetus for further increase in neuroplasticity in the hippocampus.
Collapse
Affiliation(s)
- Vadim Tseilikman
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
- Correspondence:
| | - Andrey Akulov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, 630090 Novosibirsk, Russia
| | - Oleg Shevelev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, 630090 Novosibirsk, Russia
| | - Anna Khotskina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, 630090 Novosibirsk, Russia
| | - Galina Kontsevaya
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, 630090 Novosibirsk, Russia
| | - Mikhail Moshkin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, 630090 Novosibirsk, Russia
| | - Julia Fedotova
- Laboratory of Neuroendocrinology, Pavlov Institute of Physiology, RAS, 199034 St. Petersburg, Russia
| | - Anton Pashkov
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
- FSBI “Federal Neurosurgical Center”, Nemirovich-Danchenko Str. 132/1, 630087 Novosibirsk, Russia
| | - Olga Tseilikman
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
- Department of Basic Medicine, Chelyabinsk State University, 454001 Chelyabinsk, Russia
| | - Eduard Agletdinov
- AO Vector-Best, Koltsovo Village, Research and Production Zone, Building 36, Room 211, 630559 Novosibirsk, Russia
| | - David Tseilikman
- Zelman Institute of Medicine and Psychology, Novosibirsk State University, 630090 Novosibirsk, Russia
| | | | - Evgenii Zavjalov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, 630090 Novosibirsk, Russia
| |
Collapse
|
19
|
Ketamine exerts its sustained antidepressant effects via cell-type-specific regulation of Kcnq2. Neuron 2022; 110:2283-2298.e9. [PMID: 35649415 DOI: 10.1016/j.neuron.2022.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/01/2022] [Accepted: 05/02/2022] [Indexed: 12/20/2022]
Abstract
A single sub-anesthetic dose of ketamine produces a rapid and sustained antidepressant response, yet the molecular mechanisms responsible for this remain unclear. Here, we identified cell-type-specific transcriptional signatures associated with a sustained ketamine response in mice. Most interestingly, we identified the Kcnq2 gene as an important downstream regulator of ketamine action in glutamatergic neurons of the ventral hippocampus. We validated these findings through a series of complementary molecular, electrophysiological, cellular, pharmacological, behavioral, and functional experiments. We demonstrated that adjunctive treatment with retigabine, a KCNQ activator, augments ketamine's antidepressant-like effects in mice. Intriguingly, these effects are ketamine specific, as they do not modulate a response to classical antidepressants, such as escitalopram. These findings significantly advance our understanding of the mechanisms underlying the sustained antidepressant effects of ketamine, with important clinical implications.
Collapse
|
20
|
Díaz-Hung ML, Hetz C. Proteostasis and resilience: on the interphase between individual's and intracellular stress. Trends Endocrinol Metab 2022; 33:305-317. [PMID: 35337729 DOI: 10.1016/j.tem.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 10/18/2022]
Abstract
A long proportion of the population is resilient to the negative consequences of stress. Glucocorticoids resulting from endocrine responses to stress are essential adaptive mediators, but also drive alterations to brain function, negatively impacting neuronal connectivity, synaptic plasticity, and memory-related processes. Recent evidence has indicated that organelle function and cellular stress responses are relevant determinant of vulnerability and resistance to environmental stress. At the molecular level, a fundamental mechanism of cellular stress adaptation is the maintenance of proteostasis, which also have key roles in sustaining basal neuronal function. Here, we discuss recent evidence suggesting that proteostasis unbalance at the level of the endoplasmic reticulum, the main site for protein folding in the cell, represents a possible mechanistic link between individuals and cellular stress.
Collapse
Affiliation(s)
- Mei-Li Díaz-Hung
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|
21
|
Tseilikman V, Lapshin M, Klebanov I, Chrousos G, Vasilieva M, Pashkov A, Fedotova J, Tseilikman D, Shatilov V, Manukhina E, Tseilikman O, Sarapultsev A, Downey HF. The Link between Activities of Hepatic 11beta-Hydroxysteroid Dehydrogenase-1 and Monoamine Oxidase-A in the Brain Following Repeated Predator Stress: Focus on Heightened Anxiety. Int J Mol Sci 2022; 23:ijms23094881. [PMID: 35563271 PMCID: PMC9102549 DOI: 10.3390/ijms23094881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 11/24/2022] Open
Abstract
We investigated the presence of a molecular pathway from hepatic 11-βHSD-1 to brain MAO-A in the dynamics of plasma corticosterone involvement in anxiety development. During 14 days following repeated exposure of rats to predator scent stress for 10 days, the following variables were measured: hepatic 11-βHSD-1 and brain MAO-A activities, brain norepinephrine, plasma corticosterone concentrations, and anxiety, as reflected by performance on an elevated plus maze. Anxiety briefly decreased and then increased after stress exposure. This behavioral response correlated inversely with plasma corticosterone and with brain MAO-A activity. A mathematical model described the dynamics of the biochemical variables and predicted the factor(s) responsible for the development and dynamics of anxiety. In the model, hepatic 11-βHSD-1 was considered a key factor in defining the dynamics of plasma corticosterone. In turn, plasma corticosterone and oxidation of brain ketodienes and conjugated trienes determined the dynamics of brain MAO-A activity, and MAO-A activity determined the dynamics of brain norepinephrine. Finally, plasma corticosterone was modeled as the determinant of anxiety. Solution of the model equations demonstrated that plasma corticosterone is mainly determined by the activity of hepatic 11-βHSD-1 and, most importantly, that corticosterone plays a critical role in the dynamics of anxiety following repeated stress.
Collapse
Affiliation(s)
- Vadim Tseilikman
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
- Correspondence: ; Tel.: +7-919311-85-99
| | - Maxim Lapshin
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
| | - Igor Klebanov
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
- School of Electronic Engineering and Computer Science, South Ural State University, 454080 Chelyabinsk, Russia
| | - George Chrousos
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Vasilieva
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
| | - Anton Pashkov
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
| | - Julia Fedotova
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
- Laboratory of Neuroendocrinology, Pavlov Institute of Physiology, RAS, 199034 St. Petersburg, Russia
- International Research Centre “Biotechnologies of the Third Millennium”, ITMO University, 191002 St. Petersburg, Russia
| | - David Tseilikman
- Zelman Institute of Medicine and Psychology, Novosibirsk State University, 630090 Novosibirsk, Russia;
| | - Vladislav Shatilov
- Basic Medicine Department, Chelyabinsk State University, 454001 Chelyabinsk, Russia;
| | - Eugenia Manukhina
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
- Laboratory for Regulatory Mechanisms of Stress and Adaptation, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Olga Tseilikman
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
- Basic Medicine Department, Chelyabinsk State University, 454001 Chelyabinsk, Russia;
| | - Alexey Sarapultsev
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
| | - H. Fred Downey
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (M.L.); (I.K.); (G.C.); (M.V.); (A.P.); (J.F.); (E.M.); (O.T.); (A.S.); (H.F.D.)
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
22
|
Caruso A, Ricceri L, Caruso A, Nicoletti F, Gaetano A, Scaccianoce S. Postweaning social isolation and autism-like phenotype: a biochemical and behavioral comparative analysis. Behav Brain Res 2022; 428:113891. [PMID: 35421428 DOI: 10.1016/j.bbr.2022.113891] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/15/2022] [Accepted: 04/07/2022] [Indexed: 12/16/2022]
Abstract
Adolescence is a critical period for brain development. In most mammalian species, disturbances experienced during adolescence constitute a risk factor for several neuropsychiatric disorders. In this study, we compared the biochemical and behavioral profile induced by postweaning social isolation (PWSI) in inbred C57BL/6N mice with that of BTBR mice, a rodent model of autism spectrum disorders. Male C57BL/6N mice were either housed in groups of four or isolated from weaning (postnatal day 21) for four weeks before experimental analyses. After weaning, male BTBR mice were housed four per cage and analyzed at 48 days of age. PWSI reduced hippocampal levels of type 2 metabotropic glutamate (mGlu2) receptors, and glucocorticoid and mineralocorticoid receptors. A similar reduction was seen in group-housed BTBR mice. Plasma corticosterone levels in basal conditions were not influenced by PWSI, but were increased in BTBR mice. Social investigation (total and head sniffing) and the number of ultrasonic vocalizations were reduced in both PWSI mice and age-matched group-housed BTBR mice, indicating a lower social responsiveness in both groups of mice. These results suggest that absence of social stimuli during adolescence induces an endophenotype with social deficit features, which mimics the phenotype of a mouse model of autism spectrum disorders.
Collapse
Affiliation(s)
- Alessandra Caruso
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Italy.
| | - Laura Ricceri
- Centre for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Angela Caruso
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy.
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Italy; IRCCS Neuromed, Pozzilli, Italy.
| | - Alessandra Gaetano
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Italy.
| | - Sergio Scaccianoce
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Italy.
| |
Collapse
|
23
|
Wang B, Chen D, Jiang R, Ntim M, Lu J, Xia M, Yang X, Wang Y, Kundu S, Guan R, Li S. TIP60 buffers acute stress response and depressive behaviour by controlling PPARγ-mediated transcription. Brain Behav Immun 2022; 101:410-422. [PMID: 35114329 DOI: 10.1016/j.bbi.2022.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/29/2021] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
Tat-interacting protein 60 (TIP60) as nuclear receptors (NRs) coregulator, acts as a tumor suppressor and also has promising therapeutic potential to target Alzheimer's disease. Stress has been implicated in many psychiatric disorders, and these disorders are characterized by impairments in cognitive function. Until now, there are no experimental data available on the regulatory effect of TIP60 in acute stress and depression. There is also no definitive explanation on which specific modulation of target gene expression is achieved by TIP60. Here, we identify TIP60 as a novel positive regulator in response to acute restraint stress (ARS) and a potentially effective target of antidepressants. Firstly, we discovered increased hippocampal TIP60 expressions in the ARS model. Furthermore, using the TIP60 inhibitor, MG149, we proved that TIP60 function correlates with behavioral and synaptic activation in the two-hour ARS. Secondly, the lentivirus vector (LV)-TIP60overexpression (OE) was injected into the hippocampus prior to the chronic restraint stress (CRS) experiments and it was found that over-expressed TIP60 compensates for TIP60 decrease and improves depression index in CRS. Thirdly, through the intervention of TIP60 expression in vitro, we established the genetic regulation of TIP60 on synaptic proteins, confirmed the TIP60 function as a specific coactivator for PPARγ and found that the PPARγ-mediated TIP60 function modulates transcriptional activation of synaptic proteins. Finally, the LV-TIP60OE and PPARγ antagonist, GW9662, were both administered in the CRS model and the data indicated that blocking PPARγ significantly weakened the protective effect of TIP60 against the CRS-induced depression. Conclusively, these findings together support TIP60 as a novel positive factor in response to acute stress and interacts with PPARγ to modulate the pathological mechanism of CRS-induced depression.
Collapse
Affiliation(s)
- Bin Wang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Defang Chen
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Rong Jiang
- Department of Physiology, Binzhou Medical University, Yantai Campus, 346 Guanhai Road, Laishan District, Yantai, Shandong, China
| | - Michael Ntim
- Department of Physiology, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jincheng Lu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Min Xia
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - XueWei Yang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Ying Wang
- Department of Cardiology, Institute of Heart and Vessel Diseases of Dalian Medical University, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Supratik Kundu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Rongxiao Guan
- Department of Cardiology, Institute of Heart and Vessel Diseases of Dalian Medical University, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shao Li
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
24
|
Kim W, Chung C. Brain-wide cellular mapping of acute stress-induced activation in male and female mice. FASEB J 2021; 35:e22041. [PMID: 34780680 DOI: 10.1096/fj.202101287r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/14/2021] [Accepted: 10/29/2021] [Indexed: 11/11/2022]
Abstract
Mood disorders are more prevalent and often reported to be more severe in women; however, little is known about the underlying mechanisms of this sexual prevalence. To gain insight into the functional differences in female brains in response to stress, we systemically compared brain activation in male and female C57BL/6N mice after acute stress exposure. We measured c-Fos expression levels in 18 brain areas related to stress responses after a 3-h long restraint stress and found that activation was sexually dimorphic in several brain areas, including the nucleus accumbens, ventral tegmental area, nucleus reuniens, and medial part of the lateral habenula. Moreover, stress-activated a substantial number of cells in the medial prefrontal cortex, amygdala, and lateral part of the lateral habenula; however, the levels of activation were comparable in males and females, suggesting that the core stress responding machineries are largely shared. Pearson correlation analysis revealed several interesting connections between the analyzed areas that are implicated in stress responses and depression. Overall, stress strengthened intra-circuitries in the hippocampus, amygdala, and prefrontal cortex in female mice, whereas more longer-range connections were highlighted in stressed male mice. Our study provides a highly valuable neuroanatomical framework for investigating the circuit mechanism underlying the higher vulnerability to depression in women.
Collapse
Affiliation(s)
- Woonhee Kim
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| |
Collapse
|
25
|
Karanikas E. Psychologically Traumatic Oxidative Stress; A Comprehensive Review of Redox Mechanisms and Related Inflammatory Implications. PSYCHOPHARMACOLOGY BULLETIN 2021; 51:65-86. [PMID: 34887600 PMCID: PMC8601764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The organism's energy requirements for homeostatic balance are covered by the redox mechanisms. Yet in case of psychologically traumatic stress, allostatic regulations activate both pro-oxidant and antioxidant molecules as well as respective components of the inflammatory system. Thus a new setpoint of dynamic interactions among redox elements is reached. Similarly, a multifaceted interplay between redox and inflammatory fields is activated with the mediation of major effector systems such as the immune system, Hypothalamic-Pituitary-Adrenal axis, kynurenine, and the glycaemic regulatory one. In case of sustained and/or intense traumatic stress the prophylactic antioxidant components are inadequate to provide the organism with neuroprotection finally culminating in Oxidative Stress and subsequently to cellular apoptosis. In parallel multiple inflammatory systems trigger and/or are triggered by the redox systems in tight fashion so that the causation sequence appears obscure. This exhaustive review aims at the comprehension of the interaction among components of the redox system as well as to the collection of disperse findings relative to the redox-inflammatory interplay in the context of traumatic stress so that new research strategies could be developed.
Collapse
Affiliation(s)
- Evangelos Karanikas
- Karanikas, Department of Psychiatry, General Military Hospital, Thessaloniki, Greece
| |
Collapse
|
26
|
Sturman O, von Ziegler L, Privitera M, Waag R, Duss S, Vermeiren Y, Giovagnoli L, de Deyn P, Bohacek J. Chronic adolescent stress increases exploratory behavior but does not appear to change the acute stress response in adult male C57BL/6 mice. Neurobiol Stress 2021; 15:100388. [PMID: 34527792 PMCID: PMC8430388 DOI: 10.1016/j.ynstr.2021.100388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 11/26/2022] Open
Abstract
Chronic stress exposure in adolescence can lead to a lasting change in stress responsiveness later in life and is associated with increased mental health issues in adulthood. Here we investigate whether the Chronic Social Instability (CSI) paradigm influences the behavioral and molecular responses to novel acute stressors in mice, and whether it alters physiological responses influenced by the noradrenergic system. Using large cohorts of mice, we show that CSI mice display a persistent increase in exploratory behaviors in the open field test alongside small but widespread transcriptional changes in the ventral hippocampus. However, both the transcriptomic and behavioral responses to novel acute stressors are indistinguishable between groups. In addition, the pupillometric response to a tail shock, known to be mediated by the noradrenergic system, remains unaltered in CSI mice. Ultra-high performance liquid chromatography analysis of monoaminergic neurotransmitter levels in the ventral hippocampus also shows no differences between control or CSI mice at baseline or in response to acute stress. We conclude that CSI exposure during adolescence leads to persistent changes in exploratory behavior and gene expression in the hippocampus, but it does not alter the response to acute stress in adulthood and is unlikely to alter the function of the noradrenergic system.
Collapse
Affiliation(s)
- Oliver Sturman
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Lukas von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Mattia Privitera
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Rebecca Waag
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Sian Duss
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Yannick Vermeiren
- Department of Biomedical Sciences, Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Antwerp, Belgium
- Division of Human Nutrition and Health, Chair Group of Nutritional Biology, Wageningen University & Research, Wageningen, Netherlands
- Faculty of Medicine & Health Sciences, Translational Neurosciences, University of Antwerp, Antwerp, Belgium
| | - Letizia Giovagnoli
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Peter de Deyn
- Department of Biomedical Sciences, Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen (UMCG), Groningen, Netherlands
- Department of Neurology, Memory Clinic of Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| |
Collapse
|
27
|
Tse YC, Nath M, Larosa A, Wong TP. Opposing Changes in Synaptic and Extrasynaptic N-Methyl-D-Aspartate Receptor Function in Response to Acute and Chronic Restraint Stress. Front Mol Neurosci 2021; 14:716675. [PMID: 34690693 PMCID: PMC8531402 DOI: 10.3389/fnmol.2021.716675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
A pertinent mechanism by which stress impacts learning and memory is through stress-induced plastic changes in glutamatergic transmission in the hippocampus. For instance, acute stress has been shown to alter the expression, binding, and function of the ionotropic glutamate N-methyl-D-aspartate receptor (NMDAR). However, the consequences of chronic stress, which could lead to various stress-related brain disorders, on NMDAR function remain unclear. While most studies on NMDARs focused on these receptors in synapses (synaptic NMDARs or sNMDARs), emerging findings have revealed functional roles of NMDARs outside synapses (extrasynaptic NMDARs or exNMDARs) that are distinct from those of sNMDARs. Using a restraint stress paradigm in adult rats, the objective of the current study is to examine whether sNMDARs and exNMDARs in the hippocampus are differentially regulated by acute and chronic stress. We examined sNMDAR and exNMDAR function in dorsal CA1 hippocampal neurons from brain slices of adult rats that were acutely (1 episode) or chronically (21 daily episodes) stressed by restraint (30 min). We found that acute stress increases sNMDAR but suppresses exNMDAR function. Surprisingly, we only observed a reduction in exNMDAR function after chronic stress. Taken together, our findings suggest that sNMDARs and exNMDARs may be differentially regulated by acute and chronic stress. Most importantly, the observed suppression in exNMDAR function by both acute and chronic stress implies crucial but overlooked roles of hippocampal exNMDARs in stress-related disorders.
Collapse
Affiliation(s)
- Yiu Chung Tse
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada
| | - Moushumi Nath
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Amanda Larosa
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Tak Pan Wong
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada.,Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
28
|
Glastad KM, Ju L, Berger SL. Tramtrack acts during late pupal development to direct ant caste identity. PLoS Genet 2021; 17:e1009801. [PMID: 34550980 PMCID: PMC8489709 DOI: 10.1371/journal.pgen.1009801] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/04/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
A key question in the rising field of neuroepigenetics is how behavioral plasticity is established and maintained in the developing CNS of multicellular organisms. Behavior is controlled through systemic changes in hormonal signaling, cell-specific regulation of gene expression, and changes in neuronal connections in the nervous system, however the link between these pathways is unclear. In the ant Camponotus floridanus, the epigenetic corepressor CoREST is a central player in experimentally-induced reprogramming of caste-specific behavior, from soldier (Major worker) to forager (Minor worker). Here, we show this pathway is engaged naturally on a large genomic scale during late pupal development targeting multiple genes differentially expressed between castes, and central to this mechanism is the protein tramtrack (ttk), a DNA binding partner of CoREST. Caste-specific differences in DNA binding of ttk co-binding with CoREST correlate with caste-biased gene expression both in the late pupal stage and immediately after eclosion. However, we find a unique set of exclusive Minor-bound genes that show ttk pre-binding in the late pupal stage preceding CoREST binding, followed by caste-specific gene repression on the first day of eclosion. In addition, we show that ttk binding correlates with neurogenic Notch signaling, and that specific ttk binding between castes is enriched for regulatory sites associated with hormonal function. Overall our findings elucidate a pathway of transcription factor binding leading to a repressive epigenetic axis that lies at the crux of development and hormonal signaling to define worker caste identity in C. floridanus.
Collapse
Affiliation(s)
- Karl M Glastad
- Department of Cell and Developmental Biology, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania United States of America.,Epigenetics Institute; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania United States of America
| | - Linyang Ju
- Epigenetics Institute; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania United States of America.,Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania United States of America
| | - Shelley L Berger
- Department of Cell and Developmental Biology, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania United States of America.,Epigenetics Institute; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania United States of America.,Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania United States of America
| |
Collapse
|
29
|
Sahraei M, Sahraei H, Rahimi M, Khosravi M, Ganjkhani M, Meftahi GH. Anxiogenic and anxiolytic effects of memantine injected into the ventral hippocampus in male stressed mice. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2021; 19:581-589. [PMID: 34533006 DOI: 10.1515/jcim-2021-0159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/24/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The effects of intra-ventral hippocampal memantine administration in male NMRI stressed mice were studied. METHODS Two stainless steel gauge 23 guide cannulas were placed in the middle part of the mice ventral hippocampus using stereotaxic coordination. Seven days later, the animals were undergone to the stress protocol as follows: They experience four consecutive electro-foot shock stress sessions lasting for 10 min. Five or 30 min before each stress session, the animals received intra-ventral hippocampal (0.1, 1 and, 5 µg/mouse) or intraperitoneal (1, 5, and 10 mg/kg) memantine respectively. Eight days after stress termination, the animals were tested either for the maintenance of either anxiety (elevated plus maze) or depression (forced swimming test). RESULTS Animals show anxiety eight days after stress termination. Intra-ventral hippocampal infusion of memantine (5 µg/mouse) 5 min before stress inhibited the anxiety-like behaviors. However, other doses of the drug exacerbate the stress effect. The drug, when injected peripherally exacerbated the stress effect in all doses. The drug by itself had no effect. In addition, animals also show depression nine days after stress termination and memantine (0.1, 1, and 5 µg/mouse) reduced the stress effect. The drug (0.1 µg/mouse) by itself induced depression in the animals. However, the drug when injected peripherally reduced the stress effect in all doses. CONCLUSIONS It could be concluded that NMDA glutamate receptors in the ventral hippocampus may play a pivotal role in the mediation of maintenance of anxiety and depression induced by stress in the mice.
Collapse
Affiliation(s)
- Mohammad Sahraei
- School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hedayat Sahraei
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoomeh Rahimi
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Khosravi
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mahin Ganjkhani
- Department of Physiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | |
Collapse
|
30
|
Leschik J, Lutz B, Gentile A. Stress-Related Dysfunction of Adult Hippocampal Neurogenesis-An Attempt for Understanding Resilience? Int J Mol Sci 2021; 22:7339. [PMID: 34298958 PMCID: PMC8305135 DOI: 10.3390/ijms22147339] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Newborn neurons in the adult hippocampus are regulated by many intrinsic and extrinsic cues. It is well accepted that elevated glucocorticoid levels lead to downregulation of adult neurogenesis, which this review discusses as one reason why psychiatric diseases, such as major depression, develop after long-term stress exposure. In reverse, adult neurogenesis has been suggested to protect against stress-induced major depression, and hence, could serve as a resilience mechanism. In this review, we will summarize current knowledge about the functional relation of adult neurogenesis and stress in health and disease. A special focus will lie on the mechanisms underlying the cascades of events from prolonged high glucocorticoid concentrations to reduced numbers of newborn neurons. In addition to neurotransmitter and neurotrophic factor dysregulation, these mechanisms include immunomodulatory pathways, as well as microbiota changes influencing the gut-brain axis. Finally, we discuss recent findings delineating the role of adult neurogenesis in stress resilience.
Collapse
Affiliation(s)
- Julia Leschik
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy;
| |
Collapse
|
31
|
Involvement of 5-HT1A receptor-mediated histone acetylation in the regulation of depression. Neuroreport 2021; 32:1049-1057. [PMID: 34232131 DOI: 10.1097/wnr.0000000000001693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Depression is one of the most common and disabling mental disorders. There is growing evidence that 5-HT1A receptor is involved in the regulation of depressive-related behaviors. However, the exact mechanism underlying the role of 5-HT1A receptor in depression remains unknown. Histone acetylation is associated with the pathophysiology and treatment of depression. In the current study, we investigated whether the epigenetic histone deacetylase (HDAC)-induced histone acetylation mediates the regulation of 5-HT1A receptor in depressive behaviors. We showed that 5-HT1A receptor selective agonist (±)-8-hydroxy-2-(dipropylamino) tetralin hydrobromide led to significant increase in acetylation of H3 at lysine 9 (Ac-H3K9) and H4 at lysine 5 (Ac-H4K5) and lysine 12 (Ac-H4K12) with obviously decreasing histone deacetylase 1 (HDAC1), histone deacetylase 2 (HDAC2), histone deacetylase 4 (HDAC4) and histone deacetylase 5 (HDAC5) expression in hippocampus of mice. Conversely, 5-HT1A receptor selective antagonist NAN-190 decreased the level of acetylation of H3 and H4 with increasing the expression of HDAC1, HDAC2, HDAC4 and HDAC5 in the hippocampus. Furthermore, we found that HDAC inhibitors, trichostatin A or suberoylanilide hydroxamic acid infusion to hippocampus prevented the depressive behaviors induced by NAN-190, as well as histone H3 and H4 acetylation in mice. Our results suggested that epigenetic histone acetylation coupled with 5-HT1A receptor may play vital role in the pathophysiology and treatment of depressive disorders.
Collapse
|
32
|
Wang L, Yang W, Li B, Yuan S, Wang F. Response to stress in biological disorders: Implications of stress granule assembly and function. Cell Prolif 2021; 54:e13086. [PMID: 34170048 PMCID: PMC8349659 DOI: 10.1111/cpr.13086] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 12/17/2022] Open
Abstract
It is indispensable for cells to adapt and respond to environmental stresses, in order for organisms to survive. Stress granules (SGs) are condensed membrane‐less organelles dynamically formed in the cytoplasm of eukaryotes cells to cope with diverse intracellular or extracellular stress factors, with features of liquid‐liquid phase separation. They are composed of multiple constituents, including translationally stalled mRNAs, translation initiation factors, RNA‐binding proteins and also non‐RNA‐binding proteins. SG formation is triggered by stress stimuli, viral infection and signal transduction, while aberrant assembly of SGs may contribute to tissue degenerative diseases. Recently, a growing body of evidence has emerged on SG response mechanisms for cells facing high temperatures, oxidative stress and osmotic stress. In this review, we aim to summarize factors affecting SGs assembly, present the impact of SGs on germ cell development and other biological processes. We particularly emphasize the significance of recently reported RNA modifications in SG stress responses. In parallel, we also review all current perspectives on the roles of SGs in male germ cells, with a particular focus on the dynamics of SG assembly.
Collapse
Affiliation(s)
- Lingjuan Wang
- Institute Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Weina Yang
- Institute Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Bin Li
- Tianjin Medical University General Hospital, Tianjin, China.,State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Shuiqiao Yuan
- Institute Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Fengli Wang
- Institute Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| |
Collapse
|
33
|
Oxidative Dysregulation in Early Life Stress and Posttraumatic Stress Disorder: A Comprehensive Review. Brain Sci 2021; 11:brainsci11060723. [PMID: 34072322 PMCID: PMC8228973 DOI: 10.3390/brainsci11060723] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
Traumatic stress may chronically affect master homeostatic systems at the crossroads of peripheral and central susceptibility pathways and lead to the biological embedment of trauma-related allostatic trajectories through neurobiological alterations even decades later. Lately, there has been an exponential knowledge growth concerning the effect of traumatic stress on oxidative components and redox-state homeostasis. This extensive review encompasses a detailed description of the oxidative cascade components along with their physiological and pathophysiological functions and a systematic presentation of both preclinical and clinical, genetic and epigenetic human findings on trauma-related oxidative stress (OXS), followed by a substantial synthesis of the involved oxidative cascades into specific and functional, trauma-related pathways. The bulk of the evidence suggests an imbalance of pro-/anti-oxidative mechanisms under conditions of traumatic stress, respectively leading to a systemic oxidative dysregulation accompanied by toxic oxidation byproducts. Yet, there is substantial heterogeneity in findings probably relative to confounding, trauma-related parameters, as well as to the equivocal directionality of not only the involved oxidative mechanisms but other homeostatic ones. Accordingly, we also discuss the trauma-related OXS findings within the broader spectrum of systemic interactions with other major influencing systems, such as inflammation, the hypothalamic-pituitary-adrenal axis, and the circadian system. We intend to demonstrate the inherent complexity of all the systems involved, but also put forth associated caveats in the implementation and interpretation of OXS findings in trauma-related research and promote their comprehension within a broader context.
Collapse
|
34
|
Loureiro CM, Fachim HA, Corsi-Zuelli F, Shuhama R, Menezes PR, Dalton CF, Del-Ben CM, Reynolds GP, Louzada-Junior P. The relationship of childhood trauma and DNA methylation of NMDA receptor genes in first-episode schizophrenia. Epigenomics 2021; 13:927-937. [PMID: 33942662 DOI: 10.2217/epi-2020-0451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: We investigated GRIN1, GRIN2A, GRIN2B and LINE-1 DNA methylation in first-episode schizophrenia patients, their nonaffected siblings and age- and sex-matched controls testing for associations between DNA methylation and exposition to childhood trauma. Materials & methods: The Childhood Trauma Questionnaire evaluated the history of childhood trauma. Genomic DNA was bisulfite converted and pyrosequencing was employed to quantify DNA methylation. Results: GRIN2A, GRIN2B and LINE-1 DNA methylation was not associated with childhood trauma in patients, siblings and controls. Siblings with childhood trauma had hypermethylation at CpG1 of GRIN1 compared with siblings without trauma. Conclusion: Childhood trauma may influence GRIN1 methylation in subjects with liability to psychosis, but not in frank schizophrenia or controls.
Collapse
Affiliation(s)
- Camila M Loureiro
- Department of Internal Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, 14049 9002, Brazil.,Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, S1 1WB3, UK.,Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, 14049 9004, Brazil.,Population Mental Health Center - NAP-SaMP, University of São Paulo, 01246 9035, Brazil
| | - Helene A Fachim
- Department of Endocrinology & Metabolism, Salford Royal Foundation Trust, Salford, M6 8HD6, UK
| | - Fabiana Corsi-Zuelli
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, 14049 9004, Brazil.,Department of Neurosciences & Behaviour, Ribeirão Preto Medical School, University of São Paulo, 14049 9007, Brazil
| | - Rosana Shuhama
- Population Mental Health Center - NAP-SaMP, University of São Paulo, 01246 9035, Brazil.,Department of Neurosciences & Behaviour, Ribeirão Preto Medical School, University of São Paulo, 14049 9007, Brazil
| | - Paulo R Menezes
- Population Mental Health Center - NAP-SaMP, University of São Paulo, 01246 9035, Brazil.,Department of Preventive Medicine, Faculty of Medicine, University of São Paulo, 01246 9035, Brazil
| | - Caroline F Dalton
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, S1 1WB3, UK
| | - Cristina M Del-Ben
- Population Mental Health Center - NAP-SaMP, University of São Paulo, 01246 9035, Brazil.,Department of Neurosciences & Behaviour, Ribeirão Preto Medical School, University of São Paulo, 14049 9007, Brazil
| | - Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, S1 1WB3, UK
| | - Paulo Louzada-Junior
- Department of Internal Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, 14049 9002, Brazil.,Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, 14049 9004, Brazil
| |
Collapse
|
35
|
Targeting the dysfunction of glutamate receptors for the development of novel antidepressants. Pharmacol Ther 2021; 226:107875. [PMID: 33901503 DOI: 10.1016/j.pharmthera.2021.107875] [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] [Accepted: 04/13/2021] [Indexed: 12/19/2022]
Abstract
Increasing evidence indicates that dysfunction of glutamate receptors is involved in the pathophysiology of major depressive disorder (MDD). Although accumulating efforts have been made to elucidate the applications and mechanisms underlying antidepressant-like effects of ketamine, a non-selective antagonist of N-methyl-d-aspartate receptor (NMDAR), the role of specific glutamate receptor subunit in regulating depression is not completely clear. The current review aims to discuss the relationships between glutamate receptor subunits and depressive-like behaviors. Research literatures were searched from inception to July 2020. We summarized the alterations of glutamate receptor subunits in patients with MDD and animal models of depression. Animal behaviors in response to dysfunction of glutamate receptor subunits were also surveyed. To fully understand mechanisms underlying antidepressant-like effects of modulators targeting glutamate receptors, we discussed effects of each glutamate receptor subunit on serotonin system, synaptic plasticity, neurogenesis and neuroinflammation. Finally, we collected most recent clinical applications of glutamate receptor modulators and pointed out the limitations of these candidates in the treatment of MDD.
Collapse
|
36
|
Milanesi M, Passamonti MM, Cappelli K, Minuti A, Palombo V, Sgorlon S, Capomaccio S, D’Andrea M, Trevisi E, Stefanon B, Williams JL, Ajmone-Marsan P. Genetic Regulation of Biomarkers as Stress Proxies in Dairy Cows. Genes (Basel) 2021; 12:genes12040534. [PMID: 33917627 PMCID: PMC8067459 DOI: 10.3390/genes12040534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 01/02/2023] Open
Abstract
Stress in livestock reduces productivity and is a welfare concern. At a physiological level, stress is associated with the activation of inflammatory responses and increased levels of harmful reactive oxygen species. Biomarkers that are indicative of stress could facilitate the identification of more stress-resilient animals. We examined twenty-one metabolic, immune response, and liver function biomarkers that have been associated with stress in 416 Italian Simmental and 436 Italian Holstein cows which were genotyped for 150K SNPs. Single-SNP and haplotype-based genome-wide association studies were carried out to assess whether the variation in the levels in these biomarkers is under genetic control and to identify the genomic loci involved. Significant associations were found for the plasma levels of ceruloplasmin (Bos taurus chromosome 1-BTA1), paraoxonase (BTA4) and γ-glutamyl transferase (BTA17) in the individual breed analysis that coincided with the position of the genes coding for these proteins, suggesting that their expression is under cis-regulation. A meta-analysis of both breeds identified additional significant associations with paraoxonase on BTA 16 and 26. Finding genetic associations with variations in the levels of these biomarkers suggests that the selection for high or low levels of expression could be achieved rapidly. Whether the level of expression of the biomarkers correlates with the response to stressful situations has yet to be determined.
Collapse
Affiliation(s)
- Marco Milanesi
- Department of Animal Science, Food and Nutrition—DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; (M.M.); (M.M.P.); (A.M.); (E.T.); (J.L.W.)
- Department for Innovation in Biological, Agro-Food and Forest Systems—DIBAF, Università della Tuscia, 01100 Viterbo, Italy
| | - Matilde Maria Passamonti
- Department of Animal Science, Food and Nutrition—DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; (M.M.); (M.M.P.); (A.M.); (E.T.); (J.L.W.)
| | - Katia Cappelli
- Dipartimento di Medicina Veterinaria, Università degli Studi di Perugia, 06126 Perugia, Italy; (K.C.); (S.C.)
| | - Andrea Minuti
- Department of Animal Science, Food and Nutrition—DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; (M.M.); (M.M.P.); (A.M.); (E.T.); (J.L.W.)
| | - Valentino Palombo
- Dipartimento Agricoltura Ambiente e Alimenti, Università del Molise, 86100 Campobasso, Italy; (V.P.); (M.D.)
| | - Sandy Sgorlon
- Dipartimento di Scienze Agroalimentari, Ambientali e Animali. Università degli Studi di Udine, 33100 Udine, Italy; (S.S.); (B.S.)
| | - Stefano Capomaccio
- Dipartimento di Medicina Veterinaria, Università degli Studi di Perugia, 06126 Perugia, Italy; (K.C.); (S.C.)
| | - Mariasilvia D’Andrea
- Dipartimento Agricoltura Ambiente e Alimenti, Università del Molise, 86100 Campobasso, Italy; (V.P.); (M.D.)
| | - Erminio Trevisi
- Department of Animal Science, Food and Nutrition—DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; (M.M.); (M.M.P.); (A.M.); (E.T.); (J.L.W.)
| | - Bruno Stefanon
- Dipartimento di Scienze Agroalimentari, Ambientali e Animali. Università degli Studi di Udine, 33100 Udine, Italy; (S.S.); (B.S.)
| | - John Lewis Williams
- Department of Animal Science, Food and Nutrition—DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; (M.M.); (M.M.P.); (A.M.); (E.T.); (J.L.W.)
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Adelaide, SA 5371, Australia
| | - Paolo Ajmone-Marsan
- Department of Animal Science, Food and Nutrition—DIANA, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy; (M.M.); (M.M.P.); (A.M.); (E.T.); (J.L.W.)
- Nutrigenomics and Proteomics Research Center-PRONUTRIGEN, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
- Correspondence:
| |
Collapse
|
37
|
Velli A, Iordanidou C, Asimi T, Vynichaki MI, Cholevas A, Mantouka AI, Nassens L, Chalkiadaki K, Sidiropoulou K. Sexual dimorphic effects of restraint stress on prefrontal cortical function are mediated by glucocorticoid receptor activation. Eur J Neurosci 2021; 55:2754-2765. [PMID: 33759255 DOI: 10.1111/ejn.15203] [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: 11/03/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 01/04/2023]
Abstract
Stress, a major regulator and precipitating factor of cognitive and emotional disorders, differentially manifests between males and females. Our aim was to investigate the mechanisms underlying the sexual dimorphic effects of acute restraint stress (RS) on males and females on the function of the prefrontal cortex (PFC). Adult male and female mice were subjected to RS or left in their home-cage (NR), and then tested in the light-dark test followed by the temporal order object recognition (TOR) task. Female mice exhibited increased anxiety-like levels, whereas male mice only showed deficits in the TOR task. When the behavioural tests were conducted 24 hr following restraint stress (RS24), only the reduced performance in the TOR task in male mice persisted. In a different cohort, evoked field excitatory postsynaptic potentials (fEPSPs) were recorded in layer II of acute PFC slices, immediately or 24 hr after RS. Long-term potentiation (LTP) was significantly reduced in RS and RS24 male, but not female, compared with their respective NR group. LTP in PFC slices incubated with corticosterone showed significantly reduced LTP only in males. To determine whether glucocorticoid signalling is implicated in the RS-induced behavioural effects, a different cohort of mice was administered mifepristone, a corticosterone receptor antagonist. Mifepristone administration 1 hr before RS prevented the effects of RS on the TOR task in males, but not anxiety. In conclusion, RS has differential effects on recency memory and anxiety, in males and females, which are partly mediated by the effects of corticosterone signalling on synaptic plasticity.
Collapse
Affiliation(s)
- Angeliki Velli
- Department of Biology, University of Crete, Rethimno, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklio, Greece
| | | | - Theodora Asimi
- Department of Biology, University of Crete, Rethimno, Greece
| | | | | | | | - Liesje Nassens
- Department of Biology, University of Crete, Rethimno, Greece
| | | | - Kyriaki Sidiropoulou
- Department of Biology, University of Crete, Rethimno, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklio, Greece
| |
Collapse
|
38
|
HUZARD D, RAPPENEAU V, MEIJER OC, TOUMA C, ARANGO-LIEVANO M, GARABEDIAN MJ, JEANNETEAU F. Experience and activity-dependent control of glucocorticoid receptors during the stress response in large-scale brain networks. Stress 2021; 24:130-153. [PMID: 32755268 PMCID: PMC7907260 DOI: 10.1080/10253890.2020.1806226] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The diversity of actions of the glucocorticoid stress hormones among individuals and within organs, tissues and cells is shaped by age, gender, genetics, metabolism, and the quantity of exposure. However, such factors cannot explain the heterogeneity of responses in the brain within cells of the same lineage, or similar tissue environment, or in the same individual. Here, we argue that the stress response is continuously updated by synchronized neural activity on large-scale brain networks. This occurs at the molecular, cellular and behavioral levels by crosstalk communication between activity-dependent and glucocorticoid signaling pathways, which updates the diversity of responses based on prior experience. Such a Bayesian process determines adaptation to the demands of the body and external world. We propose a framework for understanding how the diversity of glucocorticoid actions throughout brain networks is essential for supporting optimal health, while its disruption may contribute to the pathophysiology of stress-related disorders, such as major depression, and resistance to therapeutic treatments.
Collapse
Affiliation(s)
- Damien HUZARD
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Virginie RAPPENEAU
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Onno C. MEIJER
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Chadi TOUMA
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Margarita ARANGO-LIEVANO
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | | | - Freddy JEANNETEAU
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
- Corresponding author:
| |
Collapse
|
39
|
Molendijk ML, de Kloet ER. Forced swim stressor: Trends in usage and mechanistic consideration. Eur J Neurosci 2021; 55:2813-2831. [PMID: 33548153 PMCID: PMC9291081 DOI: 10.1111/ejn.15139] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/14/2021] [Accepted: 01/28/2021] [Indexed: 12/12/2022]
Abstract
The acquired immobility response during the “forced swim test (FST)” is not a rodent model of depression, but the test has some validity in predicting a compound's antidepressant potential. Nevertheless, 60% of the about 600 papers that were published annually the past 2 years label the rodent's immobility response as depression‐like behaviour, but the relative contribution per country is changing. When the Editors‐in‐Chief of 5 journals publishing most FST papers were asked for their point of view on labelling immobility as depression‐like behaviour and despair, they responded that they primarily rely on the reviewers regarding scientific merit of the submission. One Editor informs authors of the recent NIMH notice (https://grants.nih.gov/grants/guide/notice‐files/NOT‐MH‐19‐053.html) which encourages investigators to use animal models “for” addressing neurobiological questions rather than as model “of” specific mental disorders. The neurobiological questions raised by use of the FST fall in two categories. First, research on the role of endocrine and metabolic factors, with roots in the 1980s, and with focus on the bottom‐up action of glucocorticoids on circuits processing salient information, executive control and memory consolidation. Second, recent findings using novel technological and computational advances that have allowed great progress in charting top‐down control in the switch from active to passive coping with the inescapable stressor executed by neuronal ensembles of the medial prefrontal cortex via the peri‐aquaductal grey. It is expected that combining neural top‐down and endocrine bottom‐up approaches will provide new insights in the role of stress‐coping and adaptation in pathogenesis of mental disorders.
Collapse
Affiliation(s)
- Marc L Molendijk
- Institute of Psychology, Leiden University, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden University Medical Center, Leiden, The Netherlands
| | - E Ronald de Kloet
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
40
|
Elhussiny MEA, Carini G, Mingardi J, Tornese P, Sala N, Bono F, Fiorentini C, La Via L, Popoli M, Musazzi L, Barbon A. Modulation by chronic stress and ketamine of ionotropic AMPA/NMDA and metabotropic glutamate receptors in the rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110033. [PMID: 32640261 DOI: 10.1016/j.pnpbp.2020.110033] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/22/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022]
Abstract
Converging clinical and preclinical evidence has shown that dysfunction of the glutamate system is a core feature of major depressive disorder. In this context, the N-methyl-d-aspartate (NMDA) receptor antagonist ketamine has raised growing interest as fast acting antidepressant. Using the chronic mild stress (CMS) rat model of depression, performed in male rats, we aimed at analyzing whether hippocampal specific changes in subunit expression and regulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) or NMDA ionotropic receptors and in metabotropic glutamate receptors could be associated with behavioral vulnerability/resilience to CMS. We also assessed whether acute ketamine (10 mg/kg) was able to dampen the alterations in CMS vulnerable animals. Although chronic stress and ketamine had no effect on ionotropic glutamate receptors mRNAs (expression, RNA editing and splicing), we found selective modulations in their protein expression, phosphorylation and localization at synaptic membranes. AMPA GluA2 expression at synaptic membranes was significantly increased only in CMS resilient rats (although a trend was found also in vulnerable animals), while its phosphorylation at Ser880 was higher in both CMS resilient and vulnerable rats, a change partially dampened by ketamine. In the hippocampus from all stressed groups, despite NMDA receptor expression levels were reduced in total extract, the levels of GluN2B-containing NMDA receptors were remarkably increased in synaptic membranes. Finally, mGlu2 underwent a selective downregulation in stress vulnerable animals, which was completely restored by acute ketamine. Overall, these results are in line with a hypofunction of activity-dependent glutamatergic synaptic transmission induced by chronic stress exposure in all the animals, as suggested by the alterations of ionotropic glutamate receptors expression and localization at synaptic level. At the same time, the selective modulation of mGlu2 receptor, confirms its previously hypothesized functional role in regulating stress vulnerability and, for the first time here, suggests a mGlu2 involvement in the fast antidepressant effect of ketamine.
Collapse
Affiliation(s)
- Mohammed E A Elhussiny
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giulia Carini
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jessica Mingardi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Paolo Tornese
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Sezione di Fisiologia e Farmacologia, Università degli Studi di Milano, Milan, Italy
| | - Nathalie Sala
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Sezione di Fisiologia e Farmacologia, Università degli Studi di Milano, Milan, Italy
| | - Federica Bono
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Chiara Fiorentini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca La Via
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Sezione di Fisiologia e Farmacologia, Università degli Studi di Milano, Milan, Italy
| | - Laura Musazzi
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Sezione di Fisiologia e Farmacologia, Università degli Studi di Milano, Milan, Italy
| | - Alessandro Barbon
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
41
|
Unexpected role of stress as a possible resilience mechanism upon mild traumatic brain injury (mTBI) in mice. Mol Cell Neurosci 2020; 111:103586. [PMID: 33358995 DOI: 10.1016/j.mcn.2020.103586] [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: 05/07/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Mild traumatic brain injury (mTBI) is common and associated with cognitive impairment. Stress and mTBI are known to modulate the neural function. The present study aims at exploring the effect of prior stress exposure on cognitive function following mTBI. METHODS Eight weeks old male ICR mice were subjected to either stress induced by forced swimming stress alone, stress followed by an immediate mTBI, or stress followed by 30 min break and then mTBI. We had two control groups: SHAM group - a control group which was not exposed to stress nor to mTBI and control mTBI group - a control group which was exposed only to TBI with no stress. Mice were weighed prior and at 12, 24 h and 1 week following interventions. Motor evaluation was conducted by rotarod. Behavioral changes were evaluated using open field, Y maze, elevated plus maze and staircase tests, at 12 h and 1 week following interventions. Brain levels of NMDAR subunits (R1, R2A, R2B), GABABR1, glucocorticoid and mineralocorticoid receptors (GR, MR) were evaluated using western blot. RESULTS Stress alone, mTBI alone, and stress followed by immediate mTBI resulted in a significant weight loss compared to control (p < 0.05). Stress 30 min prior to mTBI had a protective effect on weight (p = 0.14 compared to control). The stress and mTBI alone groups showed reduced time at the center of the open field arena 1 week after intervention (p < 0.05 for both). Time in the novel arm of the Y maze was significantly shorter in the mTBI and stress followed by delayed mTBI (p = 0.02). Immediate stress prior to mTBI had normalized times in the novel arm (p = 0.95 compared to control). Combination of stress and mTBI significantly modified NMDAR subunits levels (increased NMDAR1, p < 0.008, decreased NMDAR2A p = 0.02) as well as increased MR levels (p = 0.04). CONCLUSION Exposure to stress prior to mTBI may improve the cognitive consequences of mTBI. These data may point towards a novel, unexpected role of stress as a possible resilience mechanism in the setting of mTBI.
Collapse
|
42
|
Fonseca R, Madeira N, Simoes C. Resilience to fear: The role of individual factors in amygdala response to stressors. Mol Cell Neurosci 2020; 110:103582. [PMID: 33346000 DOI: 10.1016/j.mcn.2020.103582] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/13/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022] Open
Abstract
Resilience to stress is an adaptive process that varies individually. Resilience refers to the adaptation, or the ability to maintain or regain mental health, despite being subject to adverse situation. Resilience is a dynamic concept that reflects a combination of internal individual factors, including age and gender interacting with external factors such as social, cultural and environmental factors. In the last decade, we have witnessed an increase in the prevalence of anxiety disorders, including post-traumatic stress disorder. Given that stress in unavoidable, it is of great interest to understand the neurophysiological mechanisms of resilience, the individual factors that may contribute to susceptibility and promote efficacious approaches to improve resilience. Here, we address this complex question, attempting at defining clear and operational definitions that may allow us to improve our analysis of behavior incorporating individuality. We examine how individual perception of the stressor can alter the outcome of an adverse situation using as an example, the fear-conditioning paradigm and discuss how individual differences in the reward system can contribute to resilience. Given the central role of the endocannabinoid system in regulating fear responses and anxiety, we discuss the evidence that polymorphisms in several molecules of this signaling system contribute to different anxiety phenotypes. The endocannabinoid system is highly interconnected with the serotoninergic and dopaminergic modulatory systems, contributing to individual differences in stress perception and coping mechanisms. We review how the individual variability in these modulatory systems can be used towards a multivariable assessment of stress risk. Incorporating individuality in our research will allow us to define biomarkers of anxiety disorders as well as assess prognosis, towards a personalized clinical approach to mental health.
Collapse
Affiliation(s)
- Rosalina Fonseca
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal.
| | - Natália Madeira
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal
| | - Carla Simoes
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal
| |
Collapse
|
43
|
Sun DS, Zhong G, Cao HX, Hu Y, Hong XY, Li T, Li X, Liu Q, Wang Q, Ke D, Liu GP, Ma RH, Luo DJ. Repeated Restraint Stress Led to Cognitive Dysfunction by NMDA Receptor-Mediated Hippocampal CA3 Dendritic Spine Impairments in Juvenile Sprague-Dawley Rats. Front Mol Neurosci 2020; 13:552787. [PMID: 33192290 PMCID: PMC7604534 DOI: 10.3389/fnmol.2020.552787] [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: 04/17/2020] [Accepted: 08/19/2020] [Indexed: 11/13/2022] Open
Abstract
Although numerous studies have indicated that chronic stress causes cognitive dysfunction with the impairment of synaptic structures and functions, the relationship between cognitive deficits induced by repeated restraint stress and the level of NMDA receptors in the subregion of the hippocampus has been relatively unknown until now. In this study, 3-week-old male Sprague-Dawley rats were exposed to repeated restraint stress for seven consecutive days, their cognitive functions were evaluated through behavioral tests, and then they were sacrificed for electrophysiological, morphological, and biochemical assays. Chronic repeated restraint stress led to cognitive and electrophysiological impairments, with a reduced density of dendritic spines. We also found that the protein level of NMDA receptors only increased in the hippocampal CA3 region. Nevertheless, repeated restraint stress-induced cognitive and synaptic dysfunction were effectively reversed by Ro25-6981, an inhibitor of the GluN2B receptor. These findings suggest that repeated restraint stress-induced synaptic and cognitive deficits are probably mediated through NMDA receptors.
Collapse
Affiliation(s)
- Dong-Sheng Sun
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Gang Zhong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Xia Cao
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Yue Hong
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Rong-Hong Ma
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan-Ju Luo
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Barfield ET, Sequeira MK, Parsons RG, Gourley SL. Morphological Responses of Excitatory Prelimbic and Orbitofrontal Cortical Neurons to Excess Corticosterone in Adolescence and Acute Stress in Adulthood. Front Neuroanat 2020; 14:45. [PMID: 33013327 PMCID: PMC7506158 DOI: 10.3389/fnana.2020.00045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/03/2020] [Indexed: 12/23/2022] Open
Abstract
Considerable evidence indicates that chronic stress and excess glucocorticoids induce neuronal remodeling in prefrontal cortical (PFC) regions. Adolescence is also characterized by a structural reorganization of PFC neurons, yet interactions between stress- and age-related structural plasticity are still being determined. We quantified dendritic spine densities on apical dendrites of excitatory neurons in the medial prefrontal cortex, prelimbic subregion (PL). Densities decreased across adolescent development, as expected, and spine volume increased. Unexpectedly, exposure to excess corticosterone (CORT) throughout adolescence did not cause additional dendritic spine loss detectable in adulthood. As a positive control dendrite population expected to be sensitive to CORT, we imaged neurons in the orbitofrontal cortex (OFC), confirming CORT-induced dendritic spine attrition on basal arbors of layer V neurons. We next assessed the effects of acute, mild stress in adulthood: On PL neurons, an acute stressor increased the density of mature, mushroom-shaped spines. Meanwhile, on OFC neurons, dendritic spine volumes and lengths were lower in mice exposed to both CORT and an acute stressor (also referred to as a "double hit"). In sum, prolonged exposure to excess glucocorticoids during adolescence can have morphological and also metaplastic consequences, but they are not global. Anatomical considerations are discussed.
Collapse
Affiliation(s)
- Elizabeth T. Barfield
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Michelle K. Sequeira
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Graduate Training Programs in Neuroscience, Emory University, Atlanta, GA, United States
| | - Ryan G. Parsons
- Graduate Program in Integrative Neuroscience, Department of Psychology, Stony Brook University, Stony Brook, NY, United States
| | - Shannon L. Gourley
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Graduate Training Programs in Neuroscience, Emory University, Atlanta, GA, United States
| |
Collapse
|
45
|
Sex-specific effects of the histone variant H2A.Z on fear memory, stress-enhanced fear learning and hypersensitivity to pain. Sci Rep 2020; 10:14331. [PMID: 32868857 PMCID: PMC7458907 DOI: 10.1038/s41598-020-71229-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/12/2020] [Indexed: 01/01/2023] Open
Abstract
Emerging evidence suggests that histone variants are novel epigenetic regulators of memory, whereby histone H2A.Z suppresses fear memory. However, it is not clear if altered fear memory can also modify risk for PTSD, and whether these effects differ in males and females. Using conditional-inducible H2A.Z knockout (cKO) mice, we showed that H2A.Z binding is higher in females and that H2A.Z cKO enhanced fear memory only in males. However, H2A.Z cKO improved memory on the non-aversive object-in-place task in both sexes, suggesting that H2A.Z suppresses non-stressful memory irrespective of sex. Given that risk for fear-related disorders, such as PTSD, is biased toward females, we examined whether H2A.Z cKO also has sex-specific effects on fear sensitization in the stress-enhanced fear learning (SEFL) model of PTSD, as well as associated changes in pain sensitivity. We found that H2A.Z cKO reduced stress-induced sensitization of fear learning and pain responses preferentially in female mice, indicating that the effects of H2A.Z depend on sex and the type of task, and are influenced by history of stress. These data suggest that H2A.Z may be a sex-specific epigenetic risk factor for PTSD susceptibility, with implications for developing sex-specific therapeutic interventions.
Collapse
|
46
|
D'Alessio L, Korman GP, Sarudiansky M, Guelman LR, Scévola L, Pastore A, Obregón A, Roldán EJA. Reducing Allostatic Load in Depression and Anxiety Disorders: Physical Activity and Yoga Practice as Add-On Therapies. Front Psychiatry 2020; 11:501. [PMID: 32581876 PMCID: PMC7287161 DOI: 10.3389/fpsyt.2020.00501] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/18/2020] [Indexed: 12/23/2022] Open
Abstract
The allostatic load (AL) index constitutes a useful tool to objectively assess the biological aspects of chronic stress in clinical practice. AL index has been positively correlated with cumulative chronic stress (physical and psychosocial stressors) and with a high risk to develop pathological conditions (e.g., metabolic syndrome, cardiovascular pathology, inflammatory disorders) and the so-called stress-related psychiatric disorders, including anxiety and depressive disorders. Chronic stress has negative effects on brain neuroplasticity, especially on hippocampal neurogenesis and these effects may be reversed by antidepressant treatments. Several evidences indicate that non-pharmacological interventions based on physical activity and yoga practice may add synergizing benefits to classical treatments (antidepressant and benzodiazepines) for depression and anxiety, reducing the negative effects of chronic stress. The aim of this review is to provide a general overview of current knowledge on AL and chronic stress in relation to depression and anxiety, physical activity and yoga practice.
Collapse
Affiliation(s)
- Luciana D'Alessio
- Universidad de Buenos Aires, Facultad de Medicina, IBCN-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Hospital Ramos Mejía, Buenos Aires, Argentina
| | - Guido Pablo Korman
- Universidad de Buenos Aires, Facultad de Psicología, CAEA-CONICET, Buenos Aires, Argentina
| | - Mercedes Sarudiansky
- Universidad de Buenos Aires, Facultad de Psicología, CAEA-CONICET, Buenos Aires, Argentina
| | - Laura Ruth Guelman
- Universidad de Buenos Aires, Facultad de Medicina, CEFYBO-CONICET, Buenos Aires, Argentina
| | - Laura Scévola
- Universidad de Buenos Aires, Hospital Ramos Mejía, Buenos Aires, Argentina
| | | | - Amilcar Obregón
- Dirección Médica y Científica, Gador SA, Buenos Aires, Argentina
| | | |
Collapse
|
47
|
Sial OK, Parise EM, Parise LF, Gnecco T, Bolaños-Guzmán CA. Ketamine: The final frontier or another depressing end? Behav Brain Res 2020; 383:112508. [PMID: 32017978 PMCID: PMC7127859 DOI: 10.1016/j.bbr.2020.112508] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 12/12/2022]
Abstract
Two decades ago, the observation of a rapid and sustained antidepressant response after ketamine administration provided an exciting new avenue in the search for more effective therapeutics for the treatment of clinical depression. Research elucidating the mechanism(s) underlying ketamine's antidepressant properties has led to the development of several hypotheses, including that of disinhibition of excitatory glutamate neurons via blockade of N-methyl-d-aspartate (NMDA) receptors. Although the prominent understanding has been that ketamine's mode of action is mediated solely via the NMDA receptor, this view has been challenged by reports implicating other glutamate receptors such as AMPA, and other neurotransmitter systems such as serotonin and opioids in the antidepressant response. The recent approval of esketamine (Spravato™) for the treatment of depression has sparked a resurgence of interest for a deeper understanding of the mechanism(s) underlying ketamine's actions and safe therapeutic use. This review aims to present our current knowledge on both NMDA and non-NMDA mechanisms implicated in ketamine's response, and addresses the controversy surrounding the antidepressant role and potency of its stereoisomers and metabolites. There is much that remains to be known about our understanding of ketamine's antidepressant properties; and although the arrival of esketamine has been received with great enthusiasm, it is now more important than ever that its mechanisms of action be fully delineated, and both the short- and long-term neurobiological/functional consequences of its treatment be thoroughly characterized.
Collapse
MESH Headings
- Antidepressive Agents/pharmacology
- Antidepressive Agents/therapeutic use
- Depressive Disorder, Major/drug therapy
- Depressive Disorder, Treatment-Resistant/drug therapy
- Dopamine Plasma Membrane Transport Proteins/drug effects
- Excitatory Amino Acid Antagonists/pharmacology
- Excitatory Amino Acid Antagonists/therapeutic use
- Humans
- Ketamine/pharmacology
- Ketamine/therapeutic use
- Norepinephrine Plasma Membrane Transport Proteins/drug effects
- Receptor, Muscarinic M1/drug effects
- Receptors, AMPA/drug effects
- Receptors, Dopamine D2/drug effects
- Receptors, N-Methyl-D-Aspartate/drug effects
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, mu/drug effects
- Receptors, Serotonin, 5-HT3/drug effects
- Receptors, sigma/drug effects
- Serotonin Plasma Membrane Transport Proteins/drug effects
Collapse
Affiliation(s)
- Omar K Sial
- Texas A&M University: Department of Psychological and Brain Sciences, 4325 TAMU, College Station, TX, 77843, USA
| | - Eric M Parise
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, 10029, USA
| | - Lyonna F Parise
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, 10029, USA
| | - Tamara Gnecco
- Texas A&M University: Department of Psychological and Brain Sciences, 4325 TAMU, College Station, TX, 77843, USA
| | - Carlos A Bolaños-Guzmán
- Texas A&M University: Department of Psychological and Brain Sciences, 4325 TAMU, College Station, TX, 77843, USA.
| |
Collapse
|
48
|
Snyder MA, Gao WJ. NMDA receptor hypofunction for schizophrenia revisited: Perspectives from epigenetic mechanisms. Schizophr Res 2020; 217:60-70. [PMID: 30979669 PMCID: PMC7258307 DOI: 10.1016/j.schres.2019.03.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023]
Abstract
Schizophrenia (SZ) is a neurodevelopmental disorder with cognitive deficits manifesting during early stages of the disease. Evidence suggests that genetic factors in combination with environmental insults lead to complex changes to glutamatergic, GABAergic, and dopaminergic systems. In particular, the N-methyl-d-aspartate receptor (NMDAR), a major glutamate receptor subtype, is implicated in both the disease progression and symptoms of SZ. NMDARs are critical for synaptic plasticity and cortical maturation, as well as learning and memory processes. In fact, any deviation from normal NMDAR expression and function can have devastating consequences. Surprisingly, there is little evidence from human patients that direct mutations of NMDAR genes contribute to SZ. One intriguing hypothesis is that epigenetic changes, which could result from early insults, alter protein expression and contribute to the NMDAR hypofunction found in SZ. Epigenetics is referred to as modifications that alter gene transcription without changing the DNA sequence itself. In this review, we first discuss how epigenetic changes to NMDAR genes could contribute to NMDAR hypofunction. We then explore how NMDAR hypofunction may contribute to epigenetic changes in other proteins or genes that lead to synaptic dysfunction and symptoms in SZ. We argue that NMDAR hypofunction occurs in early stage of the disease, and it may consequentially initiate GABA and dopamine deficits. Therefore, targeting NMDAR dysfunction during the early stages would be a promising avenue for prevention and therapeutic intervention of cognitive and social deficits that remain untreatable. Finally, we discuss potential questions regarding the epigenetic of SZ and future directions for research.
Collapse
Affiliation(s)
- Melissa A. Snyder
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada, K1H 8M5,Correspondence: Wen-Jun Gao, M.D., Ph.D., Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, Phone: (215) 991-8907, Fax: (215) 843-9802, ; Melissa A. Snyder, Ph.D.,
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America.
| |
Collapse
|
49
|
Wilson DF, Matschinsky FM. Ethanol metabolism: The good, the bad, and the ugly. Med Hypotheses 2020; 140:109638. [PMID: 32113062 DOI: 10.1016/j.mehy.2020.109638] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/20/2022]
Abstract
Throughout the world, ethanol is both an important commercial commodity and a source of major medical and social problems. Ethanol readily passes through biological membranes and distributes throughout the body. It is oxidized, first to acetaldehyde and then to acetate, and finally by the citric acid cycle in virtually all tissues. The oxidation of ethanol is irreversible and unregulated, making the rate dependent only on local concentration and enzyme activity. This unregulated input of reducing equivalents increases reduction of both cytoplasmic and intramitochondrial NAD and, through the latter, cellular energy state {[ATP]/([ADP][Pi])}. In brain, this increase in energy state stimulates dopaminergic neural activity signalling reward and a sense of well being, while suppressing glutamatergic neural activity signalling anxiety and unease. These positive responses to ethanol ingestion are important to social alcohol consumption. Importantly, decreased free [AMP] decreases AMP-dependent protein kinase (AMPK) activity, an important regulator of cellular energy metabolism. Oxidation of substrates used for energy metabolism in the absence of ethanol is down regulated to accommodate the input from ethanol. In liver, chronic ethanol metabolism results in fatty liver and general metabolic dysfunction. In brain, transport of other oxidizable metabolites through the blood-brain barrier and the enzymes for their oxidation are both down regulated. For exposures of short duration, ethanol induced regulatory changes are rapid and reversible, recovering completely when the concentrations of ethanol and acetate fall again. Longer periods of ethanol exposure and associated chronic suppression of AMPK activity activates regulatory mechanisms, including gene expression, that operate over longer time scales, both in onset and reversal. If chronic alcohol consumption is abruptly ended, metabolism is no longer able to respond rapidly enough to compensate. Glutamatergic neural activity adapts to chronic dysregulation of glutamate metabolism and suppression of glutamatergic neural activity by increasing excitatory and decreasing inhibitory amino acid receptors. A point is reached (ethanol dependence) where withdrawal of ethanol results in significant metabolic energy depletion in neurons and other brain cells as well as hyperexcitation of the glutamatergic system. The extent and regional specificity of energy depletion in the brain, combined with hyperactivity of the glutamatergic neuronal system, largely determines the severity of withdrawal symptoms.
Collapse
Affiliation(s)
- David F Wilson
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Franz M Matschinsky
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
50
|
Coffman JA. Chronic stress, physiological adaptation and developmental programming of the neuroendocrine stress system. FUTURE NEUROLOGY 2020. [DOI: 10.2217/fnl-2019-0014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Chronic stress undermines physical and mental health, in part via dysregulation of the neuroendocrine stress system. Key to understand this dysregulation is recognizing that the problem is not stress per se, but rather its chronicity. The optimally functioning stress system is highly dynamic, and negative feedback regulation enforces transient responses to acute stressors. Chronic stress overrides this, and adaptation to the chronicity can result in persistent dysregulation by altering sensitivity thresholds critical for control of system dynamics. Such adaptation involves plasticity within the central nervous system (CNS) as well as epigenetic regulation. When it occurs during development, it can have persistent effects on neuroendocrine regulation. Understanding how chronic stress programs development of the neuroendocrine stress system requires elucidation of stress-responsive gene regulatory networks that control CNS plasticity and development.
Collapse
Affiliation(s)
- James A Coffman
- MDI Biological Laboratory, Kathryn W Davis Center for Regenerative Biology and Aging, Salisbury Cove, ME 04672, USA
| |
Collapse
|