1
|
Lugenbühl JF, Viho EMG, Binder EB, Daskalakis NP. Stress Molecular Signaling in Interaction With Cognition. Biol Psychiatry 2025; 97:349-358. [PMID: 39368530 PMCID: PMC11896655 DOI: 10.1016/j.biopsych.2024.09.023] [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: 06/02/2024] [Revised: 09/02/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
Exposure to stressful life events is associated with a high risk of developing psychiatric disorders with a wide variety of symptoms. Cognitive symptoms in stress-related psychiatric disorders can be particularly challenging to understand, both for those experiencing them and for health care providers. To gain insights, it is important to capture stress-induced structural, epigenomic, transcriptomic, and proteomic changes in relevant brain regions such as the amygdala, hippocampus, locus coeruleus, and prefrontal cortex that result in long-lasting alterations in brain function. In this review, we will emphasize a subset of stress molecular mechanisms that alter neuroplasticity, neurogenesis, and balance between excitatory and inhibitory neurons. Then, we discuss how to identify genetic risk factors that may accelerate stress-driven or stress-induced cognitive impairment. Despite the development of new technologies such as single-cell resolution sequencing, our understanding of the molecular effects of stress in the brain remains to be deepened. A better understanding of the diversity of stress effects in different brain regions and cell types is a prerequisite to open new avenues for mechanism-informed prevention and treatment of stress-related cognitive symptoms.
Collapse
Affiliation(s)
- Justina F Lugenbühl
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Department of Psychiatry and Neuropsychology, School for Mental Health, and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Eva M G Viho
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
| | - Nikolaos P Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
2
|
Lai W, Zhang H, Jiang Y, Huang X, Liao Y, Hao J, Liu Y, Chen Y, Wang W, Guo L, Zhang WH, Teopiz KM, McIntyre RS, Han X, Lu C. Trauma exposure as a risk factor of major depressive disorder: A matched case-control study. J Psychiatr Res 2025; 182:204-211. [PMID: 39818108 DOI: 10.1016/j.jpsychires.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
The association between childhood trauma (CT), stressful life events (SLE) and the onset and severity of major depressive disorder (MDD) has not been extensively studied. This study aimed to investigate the separate and combined association of CT and SLE with the onset and severity of MDD. A total of 503 patients with MDD and 503 controls were included. The diagnosis of MDD was assessed using the Mini-International Neuropsychiatric Interview (M.I.N.I.) by trained psychiatrists. Trauma exposure was assessed using the short form of Childhood Trauma Questionnaire (CTQ-SF) and stressful life events screening questionnaire (SLESQ). A series of conditional logistic regression models and multiple linear regression models were performed. The majority of patients with MDD (84.9%) and controls (61.6%) have reported trauma exposure. After adjusting for covariates, exposure to CT or SLE was independently associated with an elevated risk of the onset and severity of MDD. Moreover, a significant additive interaction between CT and SLE on the onset of MDD was observed (relative excess risk due to interactions [RERI] = 6.93, 95% confidence interval [CI] = 0.19-13.66). MDD patients were more likely to experience both CT and SLE compared with controls (odds ratio [OR] = 10.37, 95% CI = 4.99-21.56). MDD patients with both CT and SLE experience were more like to have more severe depressive symptoms (β = 2.16, 95% CI = 1.02-3.30). These findings underscore the importance of targeted prevention measures addressing the occurrence of CT and SLE to mitigate the risk of MDD development and the severity of depressive symptoms.
Collapse
Affiliation(s)
- Wenjian Lai
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen university, 510080, Guangzhou, China; Guangdong Engineering Technology Research Center of Nutrition Translation, 510080, Guangzhou, China; International Centre for Reproductive Health (ICRH), Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Huimin Zhang
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Yingchen Jiang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen university, 510080, Guangzhou, China; Guangdong Engineering Technology Research Center of Nutrition Translation, 510080, Guangzhou, China
| | - Xinyu Huang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen university, 510080, Guangzhou, China; Guangdong Engineering Technology Research Center of Nutrition Translation, 510080, Guangzhou, China
| | - Yuhua Liao
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Jiejing Hao
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen university, 510080, Guangzhou, China; Guangdong Engineering Technology Research Center of Nutrition Translation, 510080, Guangzhou, China; Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Yifen Liu
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Yan Chen
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Wanxin Wang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen university, 510080, Guangzhou, China; Guangdong Engineering Technology Research Center of Nutrition Translation, 510080, Guangzhou, China
| | - Lan Guo
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen university, 510080, Guangzhou, China; Guangdong Engineering Technology Research Center of Nutrition Translation, 510080, Guangzhou, China
| | - Wei-Hong Zhang
- International Centre for Reproductive Health (ICRH), Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; School of Public Health, Université libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Kayla M Teopiz
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Roger S McIntyre
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Xue Han
- Department of Psychiatry, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China.
| | - Ciyong Lu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen university, 510080, Guangzhou, China; Guangdong Engineering Technology Research Center of Nutrition Translation, 510080, Guangzhou, China.
| |
Collapse
|
3
|
Tian X, Russo SJ, Li L. Behavioral Animal Models and Neural-Circuit Framework of Depressive Disorder. Neurosci Bull 2025; 41:272-288. [PMID: 39120643 PMCID: PMC11794861 DOI: 10.1007/s12264-024-01270-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/26/2024] [Indexed: 08/10/2024] Open
Abstract
Depressive disorder is a chronic, recurring, and potentially life-endangering neuropsychiatric disease. According to a report by the World Health Organization, the global population suffering from depression is experiencing a significant annual increase. Despite its prevalence and considerable impact on people, little is known about its pathogenesis. One major reason is the scarcity of reliable animal models due to the absence of consensus on the pathology and etiology of depression. Furthermore, the neural circuit mechanism of depression induced by various factors is particularly complex. Considering the variability in depressive behavior patterns and neurobiological mechanisms among different animal models of depression, a comparison between the neural circuits of depression induced by various factors is essential for its treatment. In this review, we mainly summarize the most widely used behavioral animal models and neural circuits under different triggers of depression, aiming to provide a theoretical basis for depression prevention.
Collapse
Affiliation(s)
- Xiangyun Tian
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Long Li
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
4
|
Rozov A, Fedulina A, Krut’ V, Sokolov R, Sulimova A, Jappy D. Influence of early-life stress on hippocampal synaptic and network properties. Front Neural Circuits 2024; 18:1509254. [PMID: 39749113 PMCID: PMC11693662 DOI: 10.3389/fncir.2024.1509254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025] Open
Abstract
According to the World Health Organization, the number of people suffering from depressive disorders worldwide is approaching 350 million. The consequences of depressive disorders include considerable worsening of the quality of life, which frequently leads to social isolation. One of the key factors which may cause depression in adulthood is early life stress, in particular, insufficient maternal care during infancy. Studies performed with children raised in orphanages have shown that long-term complete absence of maternal care (chronic early life stress) leads to vulnerability to emotional disorders, including depression, in adulthood. All of the above dictates the need for a deep understanding of the mechanisms of the pathogenicity of stress in neurogenesis. Therefore, the consequences of stress experienced in the early stages of development are actively studied in animal models. A large body of evidence has accumulated indicating stress-induced changes in gene expression and behavioral disorders in adulthood. However, the connection between the molecular biology of neurons and complex behavior runs through the synaptic connections linking these neurons into complex neural networks. In turn, coordinated activity in neuronal ensembles, achieved by a balance of synaptic excitation and inhibition, is the basis of complex behavior. Unfortunately, the effect of stress on synaptic interactions of neurons remains poorly understood.
Collapse
Affiliation(s)
- Andrei Rozov
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | - Anastasia Fedulina
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Viktoriya Krut’
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | - Rostislav Sokolov
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Arina Sulimova
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | - David Jappy
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
5
|
San Felipe D, Martín-Sánchez B, Zekri-Nechar K, Moya M, Llorente R, Zamorano-León JJ, Marco EM, López-Gallardo M. Consequences of Early Maternal Deprivation on Neuroinflammation and Mitochondrial Dynamics in the Central Nervous System of Male and Female Rats. BIOLOGY 2024; 13:1011. [PMID: 39765678 PMCID: PMC11672930 DOI: 10.3390/biology13121011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025]
Abstract
Early life stress (ELS) is associated with an increased risk for neuropsychiatric disorders, and both neuroinflammation and mitochondrial dysfunction seem to be central to mental health. Herein, using an animal model of ELS, a single episode of maternal deprivation (MD, 24 h on pnd 9) extensively documented to elicit behavioural anomalies in male and female Wistar rats, we investigated its consequences in terms of neuroinflammation and mitochondrial dynamics in the prefrontal cortex (PFC) and the hippocampal formation (HCF). MD differentially affected the brain content of cytokines: MD induced a transient increase in pro-inflammatory cytokines (IL-1β and IL-6) in the PFC, as well as in the levels of the anti-inflammatory cytokine IL-10 in the HCF. MD also induced a significant decrease mitochondria citrate synthase activity, but MD did not exert significant changes in mitochondria Complex IV activity, revealing a generalized decrease in mitochondrial density without any change in mitochondrial respiration. In the present study, we demonstrate that MD induces neuroinflammatory processes in specific brain regions. Additional research is needed to better understand the temporal pattern of such changes, their impact on the developing brain, and their participation in the already well-known behavioural consequences of MD.
Collapse
Affiliation(s)
- Diego San Felipe
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| | - Beatriz Martín-Sánchez
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| | - Khaoula Zekri-Nechar
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Department of Public Health and Maternal-Child Health, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain;
| | - Marta Moya
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| | - Ricardo Llorente
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| | - Jose J. Zamorano-León
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Department of Public Health and Maternal-Child Health, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain;
| | - Eva M. Marco
- Department of Genetics, Physiology and Microbiology, Faculty of Biological Sciences, Complutense University of Madrid, C/José Antonio Novais 12, 28040 Madrid, Spain
| | - Meritxell López-Gallardo
- Department of Physiology, School of Medicine, Complutense University of Madrid, Pza. Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (D.S.F.); (B.M.-S.); (M.M.); (R.L.); (M.L.-G.)
| |
Collapse
|
6
|
de la Puente-Aldea J, Lopez-Llanos O, Horrillo D, Marcos-Sanchez H, Sanz-Ballesteros S, Franco R, Jaisser F, Senovilla L, Palacios-Ramirez R. Mineralocorticoid Receptor and Sleep Quality in Chronic Kidney Disease. Int J Mol Sci 2024; 25:12320. [PMID: 39596384 PMCID: PMC11594958 DOI: 10.3390/ijms252212320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
The classical function of the mineralocorticoid receptor (MR) is to maintain electrolytic homeostasis and control extracellular volume and blood pressure. The MR is expressed in the central nervous system (CNS) and is involved in the regulation of the hypothalamic-pituitary-adrenal (HPA) axis as well as sleep physiology, playing a role in the non-rapid eye movement (NREM) phase of sleep. Some patients with psychiatric disorders have very poor sleep quality, and a relationship between MR dysregulation and this disorder has been found in them. In addition, the MR is involved in the regulation of the renal peripheral clock. One of the most common comorbidities observed in patients with chronic kidney disease (CKD) is poor sleep quality. Patients with CKD experience sleep disturbances, including reduced sleep duration, sleep fragmentation, and insomnia. To date, no studies have specifically investigated the relationship between MR activation and CKD-associated sleep disturbances. However, in this review, we analyzed the environment that occurs in CKD and proposed two MR-related mechanisms that may be responsible for these sleep disturbances: the circadian clock disruption and the high levels of MR agonist observed in CKD.
Collapse
Affiliation(s)
- Juan de la Puente-Aldea
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid—CSIC, 47003 Valladolid, Spain; (J.d.l.P.-A.); (O.L.-L.); (L.S.)
| | - Oscar Lopez-Llanos
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid—CSIC, 47003 Valladolid, Spain; (J.d.l.P.-A.); (O.L.-L.); (L.S.)
| | - Daniel Horrillo
- Facultad de ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcon, Spain; (D.H.); (R.F.)
| | | | | | - Raquel Franco
- Facultad de ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcon, Spain; (D.H.); (R.F.)
| | - Frederic Jaisser
- INSERM U1166, Team Diabetes, Metabolic Diseases and Comorbidities, Sorbonne Université, 75013 Paris, France;
- INSERM UMR 1116, Centre d’Investigations Cliniques-Plurithématique 1433, Université de Lorraine, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, 54500 Nancy, France
| | - Laura Senovilla
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid—CSIC, 47003 Valladolid, Spain; (J.d.l.P.-A.); (O.L.-L.); (L.S.)
- INSERM U1138, Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Sorbonne Université, Institut Universitaire de France, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
| | - Roberto Palacios-Ramirez
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid—CSIC, 47003 Valladolid, Spain; (J.d.l.P.-A.); (O.L.-L.); (L.S.)
| |
Collapse
|
7
|
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress / adversity: Structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress 2024; 33:100669. [PMID: 39309367 PMCID: PMC11415888 DOI: 10.1016/j.ynstr.2024.100669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Adverse early life experiences are strongly associated with reduced cognitive function throughout life. The link is strong in many human studies, but these do not enable assigning causality, and the limited access to the live human brain can impede establishing the mechanisms by which early-life adversity (ELA) may induce cognitive problems. In experimental models, artificially imposed chronic ELA/stress results in deficits in hippocampus dependent memory as well as increased vulnerability to the deleterious effects of adult stress on memory. This causal relation of ELA and life-long memory impairments provides a framework to probe the mechanisms by which ELA may lead to human cognitive problems. Here we focus on the consequences of a one-week exposure to adversity during early postnatal life in the rodent, the spectrum of the ensuing memory deficits, and the mechanisms responsible. We highlight molecular, cellular and circuit mechanisms using convergent trans-disciplinary approaches aiming to enable translation of the discoveries in experimental models to the clinic.
Collapse
Affiliation(s)
- Tallie Z. Baram
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| | - Matthew T. Birnie
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
8
|
Koulouri A, Zannas AS. Epigenetics as a link between environmental factors and dementia risk. J Alzheimers Dis Rep 2024; 8:1372-1380. [PMID: 40034348 PMCID: PMC11863733 DOI: 10.1177/25424823241284227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/27/2024] [Indexed: 03/05/2025] Open
Abstract
Dementia encompasses a broad spectrum of neuropsychiatric disease states marked by cognitive impairments that interfere with day-to-day functioning. Most dementias are complex phenotypes that result from a genome-environment interplay. Epigenetic regulation has emerged as a candidate mechanism for studying this interplay. In this narrative review, we discuss state-of-the-art evidence on environmental exposures relevant to dementia, including nutrition, physical exercise, psychosocial stress, and environmental toxins, and highlight epigenetic mechanisms that have been reported as a putative link between each exposure and dementia risk. We then discuss the clinical implications and future directions of this line of research. An improved understanding of the epigenetic mechanisms involved in dementia pathogenesis can promote the development of novel biomarkers for predicting outcomes but also targeted therapies to intervene early in the course of the disease.
Collapse
Affiliation(s)
- Adamantia Koulouri
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Anthony S Zannas
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
9
|
Dixon R, Malave L, Thompson R, Wu S, Li Y, Sadik N, Anacker C. Sex-specific and Developmental Effects of Early Life Adversity on Stress Reactivity are Rescued by Postnatal Knockdown of 5-HT 1A Autoreceptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576344. [PMID: 38328253 PMCID: PMC10849559 DOI: 10.1101/2024.01.22.576344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Early Life Adversity (ELA) predisposes to stress hypersensitivity in adulthood, but neurobiological mechanisms that protect from the enduring effects of ELA are poorly understood. Serotonin 1A (5HT 1A ) autoreceptors in the raphé nuclei regulate adult stress vulnerability, but whether 5HT 1A could be targeted to prevent ELA effects on susceptibility to future stressors is unknown. Here, we exposed mice with postnatal knockdown of 5HT 1A autoreceptors to the limited bedding and nesting model of ELA from postnatal day (P)3-10 and tested behavioral, neuroendocrine, neurogenic, and neuroinflammatory responses to an acute swim stress in male and female mice in adolescence (P35) and in adulthood (P56). In females, ELA decreased raphé 5HT neuron activity in adulthood and increased passive coping with the acute swim stress, corticosterone levels, neuronal activity, and corticotropin-releasing factor (CRF) levels in the paraventricular nucleus (PVN) of the hypothalamus. ELA also reduced neurogenesis in the ventral dentate gyrus (vDG) of the hippocampus, an important mediator of individual differences in stress susceptibility, and increased microglia activation in the PVN and vDG. These effects of ELA were specific to females and manifested predominantly in adulthood, but not earlier on in adolescence. Postnatal knockdown of 5HT 1A autoreceptors prevented these effects of ELA on 5HT neuron activity, stress reactivity, neurogenesis, and neuroinflammation in adult female mice. Our findings demonstrate that ELA induces long-lasting and sex-specific impairments in the serotonin system, stress reactivity, and vDG function, and identify 5HT 1A autoreceptors as potential targets to prevent these enduring effects of ELA.
Collapse
|
10
|
Theleritis C, Stefanou MI, Demetriou M, Alevyzakis E, Triantafyllou K, Smyrnis N, Spandidos DA, Rizos E. Association of gut dysbiosis with first‑episode psychosis (Review). Mol Med Rep 2024; 30:130. [PMID: 38785152 PMCID: PMC11148526 DOI: 10.3892/mmr.2024.13254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
The gut‑microbiota‑brain axis is a complex bidirectional communication system linking the gastrointestinal tract to the brain. Changes in the balance, composition and diversity of the gut‑microbiota (gut dysbiosis) have been found to be associated with the development of psychosis. Early‑life stress, along with various stressors encountered in different developmental phases, have been shown to be associated with the abnormal composition of the gut microbiota, leading to irregular immunological and neuroendocrine functions, which are potentially responsible for the occurrence of first‑episode psychosis (FEP). The aim of the present narrative review was to summarize the significant differences of the altered microbiome composition in patients suffering from FEP vs. healthy controls, and to discuss its effects on the occurrence and intensity of symptoms in FEP.
Collapse
Affiliation(s)
- Christos Theleritis
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Maria-Ioanna Stefanou
- Second Department of Neurology, School of Medicine, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Marina Demetriou
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Evangelos Alevyzakis
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, Second Department of Propaedeutic Internal Medicine, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Nikolaos Smyrnis
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Emmanouil Rizos
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
11
|
Cattane N, Di Benedetto MG, D'Aprile I, Riva MA, Cattaneo A. Dissecting the Long-Term Effect of Stress Early in Life on FKBP5: The Role of miR-20b-5p and miR-29c-3p. Biomolecules 2024; 14:371. [PMID: 38540789 PMCID: PMC10967956 DOI: 10.3390/biom14030371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024] Open
Abstract
Exposure to early-life stress (ELS) has been related to an increased susceptibility to psychiatric disorders later in life. Although the molecular mechanisms underlying this association are still under investigation, glucocorticoid signaling has been proposed to be a key mediator. Here, we used two preclinical models, the prenatal stress (PNS) animal model and an in vitro model of hippocampal progenitor cells, to assess the long-term effect of ELS on FKBP5, NR3C1, NR3C2, and FoxO1, four stress-responsive genes involved in the effects of glucocorticoids. In the hippocampus of male PNS rats sacrificed at different time points during neurodevelopment (PND 21, 40, 62), we found a statistically significant up-regulation of FKBP5 at PND 40 and PND 62 and a significant increase in FoxO1 at PND 62. Interestingly, all four genes were significantly up-regulated in differentiated cells treated with cortisol during cell proliferation. As FKBP5 was consistently modulated by PNS at adolescence (PND 40) and adulthood (PND 62) and by cortisol treatment after cell differentiation, we measured a panel of miRNAs targeting FKBP5 in the same samples where FKBP5 expression levels were available. Interestingly, both miR-20b-5p and miR-29c-3p were significantly reduced in PNS-exposed animals (both at PND40 and 62) and also in the in vitro model after cortisol exposure. Our results highlight the key role of miR-20b-5p and miR-29c-3p in sustaining the long-term effects of ELS on the stress response system, representing a mechanistic link possibly contributing to the enhanced stress-related vulnerability to mental disorders.
Collapse
Affiliation(s)
- Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Maria Grazia Di Benedetto
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Ilari D'Aprile
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Marco Andrea Riva
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| |
Collapse
|
12
|
Gorthy AS, Balleste AF, Placeres-Uray F, Atkins CM. Chronic Stress in Early Development and Effects on Traumatic Brain Injury Outcome. ADVANCES IN NEUROBIOLOGY 2024; 42:179-204. [PMID: 39432043 PMCID: PMC11556197 DOI: 10.1007/978-3-031-69832-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
In recent years, significant advances have been made in the study of mild traumatic brain injury (mTBI). Complete recovery from mTBI normally requires days to weeks, yet a subset of the population suffers from symptoms for weeks to months after injury. The risk factors for these prolonged symptoms have not yet been fully understood. In this chapter, we address one proposed risk factor, early life stress (ELS) and its influence on mTBI recovery. To study the effects of ELS on mTBI recovery, accepted animal models of ELS, including maternal separation, limited bedding and nesting, and chronic unpredictable stress, have been implemented. Combining these ELS models with standardized mTBI models, such as fluid percussion injury or controlled cortical impact, has allowed for a deeper understanding of the neuronal, hormonal, and cognitive changes that occur after mTBI following ELS. These preclinical findings are being used to understand how adverse childhood experiences may predispose a subset of individuals to poorer recovery after mTBI.
Collapse
Affiliation(s)
- Aditi S Gorthy
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alyssa F Balleste
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fabiola Placeres-Uray
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Coleen M Atkins
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
13
|
Bagheri F, Goudarzi I, Lashkarbolouki T, Elahdadi Salmani M, Goudarzi A, Morley-Fletcher S. Improving behavioral deficits induced by perinatal ethanol and stress exposure in adolescent male rat progeny via maternal melatonin treatment. Psychopharmacology (Berl) 2024; 241:153-169. [PMID: 37889278 DOI: 10.1007/s00213-023-06470-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND AND AIM Early-life stressful situations and binge drinking have been thus far acknowledged as two burdensome conditions that potentially give rise to negative outcomes and then synergistically affect brain development. In this context, the hippocampus, with the greatest number of glucocorticoid receptors (GCRs) in the brain, is responsible for regulating negative responses to stress. Prolonged glucocorticoid (GC) exposure can accordingly cause oxidative stress (OS), leading to cognitive and emotional dysfunction. Against this background, melatonin, as a powerful antioxidant and hypothalamus-pituitary-adrenal (HPA) axis regulator, was administered in this study to ameliorate cognitive impairments induced by perinatal ethanol and stress exposure in adolescent male rat progeny. METHODS Wistar rat dams were exposed to ethanol (4 g/kg) and melatonin (10 mg/kg) from gestational day (GD) 6 to postnatal day (PND) 14 and then limited nesting material (LNS) from PND0 to PND14 individually or in combination. Maternal behavior was then investigated in mothers. Afterward, the plasma corticosterone (CORT) concentration, the OS marker, the corticotropin-releasing hormone receptor type 1 (CRHR1) expression, and the GCR and brain-derived neurotrophic factor (BDNF) levels were measured in the male pups. Moreover, behavioral tasks, including the elevated plus maze (EPM), the Morris water maze (MWM), the novel object recognition (NORT), and the object-location memory (OLM) tests were completed and assessed. RESULTS The quantity and quality of maternal care significantly decreased in the mothers with dual exposure to ethanol and stress. The plasma CORT concentration in the progeny also dropped in the Ethanol + LNS group, but the risk-taking behavior elevated significantly. The ethanol and stress exposure further revealed a significant fall in the GCR and CRHR1 expression levels, compared with stress alone. The results of learning and memory tasks also indicated a significant reduction in spatial learning and memory among animals exposed to ethanol and stress. The BDNF mRNA levels correspondingly increased in the Ethanol + LNS group, compared with LNS alone. In the presence of ethanol and stress, the superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities correspondingly declined. On the other hand, the malondialdehyde (MDA) levels augmented in the hippocampus of the animals with ethanol and LNS dual exposure, as compared with the control group. Melatonin treatment (MT) thus improved nursing behaviors in dams, prevented OS, enhanced the CRHR1 and GCR expression, and reduced the BDNF levels to the similar ones in the control group. The animals in the Ethanol + LNS + MT group ultimately showed an ameliorated performance at behavioral tasks, including the memory and risk-taking behavior. CONCLUSION It was concluded that MT could prevent stress response and memory impairments arising from dual exposure to ethanol and stress by inhibiting OS.
Collapse
Affiliation(s)
| | - Iran Goudarzi
- School of Biology, Damghan University, Damghan, Iran.
| | | | | | - Afsaneh Goudarzi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Morley-Fletcher
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale Et Fonctionnelle, 59000, Lille, France
| |
Collapse
|
14
|
Giannopoulou I, Georgiades S, Stefanou MI, Spandidos DA, Rizos E. Links between trauma and psychosis (Review). Exp Ther Med 2023; 26:386. [PMID: 37456168 PMCID: PMC10347243 DOI: 10.3892/etm.2023.12085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
The relationship between trauma and psychosis is complex and multifaceted, with evidence suggesting that trauma can be both a risk factor for the development of psychosis and a consequence of psychotic experiences. The present review aimed to provide an overview of the current state of knowledge on the relationship between trauma and psychosis, including historical and conceptual considerations, as well as epidemiological evidence. The potential explanation of the link between trauma and psychosis is provided through available models and similarities in their neurobiological associations. Overall, the research confirms the relationship between trauma and psychosis, and suggests that individuals with a co-occurring history of trauma and psychosis may have increased symptom severity and worse functional outcomes compared with individuals with psychosis alone. Future research should focus on elucidating the underlying causal pathways between trauma exposure and psychosis in order to inform effective treatment approaches aiming to prevent the intensification of psychotic symptoms and processes.
Collapse
Affiliation(s)
- Ioanna Giannopoulou
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Stelios Georgiades
- Department of Basic Clinical Sciences, Medical School, University of Nicosia, 2415 Nicosia, Cyprus
| | - Maria-Ioanna Stefanou
- Second Department of Neurology, School of Medicine, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Emmanouil Rizos
- Second Department of Psychiatry, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
15
|
Premoli M, Fyke W, Bellocchio L, Lemaire V, Wolley-Roberts M, Bontempi B, Pietropaolo S. Early Administration of the Phytocannabinoid Cannabidivarin Prevents the Neurobehavioral Abnormalities Associated with the Fmr1-KO Mouse Model of Fragile X Syndrome. Cells 2023; 12:1927. [PMID: 37566006 PMCID: PMC10416983 DOI: 10.3390/cells12151927] [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: 06/17/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023] Open
Abstract
Phytocannabinoids, including the non-addictive cannabis component cannabidivarin (CBDV), have been reported to hold therapeutic potential in several neurodevelopmental disorders (NDDs). Nonetheless, the therapeutic value of phytocannabinoids for treating Fragile X syndrome (FXS), a major NDD, remains unexplored. Here, we characterized the neurobehavioral effects of CBDV at doses of 20 or 100 mg/kg in the Fmr1-knockout (Fmr1-KO) mouse model of FXS using two temporally different intraperitoneal regimens: subchronic 10-day delivery during adulthood (Study 1: rescue treatment) or chronic 5-week delivery at adolescence (Study 2: preventive treatment). Behavioral tests assessing FXS-like abnormalities included anxiety, locomotor, cognitive, social and sensory alterations. Expression of inflammatory and plasticity markers was investigated in the hippocampus and prefrontal cortex. When administered during adulthood (Study 1), the effects of CBDV were marginal, rescuing at the lower dose only the acoustic hyper-responsiveness of Fmr1-KO mice and at both doses their altered hippocampal expression of neurotrophins. When administered during adolescence (Study 2), CBDV at both doses prevented the cognitive, social and acoustic alterations of adult Fmr1-KO mice and modified the expression of several inflammatory brain markers in both wild-type littermates and mutants. These findings warrant the therapeutic potential of CBDV for preventing neurobehavioral alterations associated with FXS, highlighting the relevance of its early administration.
Collapse
Affiliation(s)
- Marika Premoli
- CNRS, EPHE, INCIA, UMR 5287, Univ. Bordeaux, 33000 Bordeaux, France
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - William Fyke
- CNRS, EPHE, INCIA, UMR 5287, Univ. Bordeaux, 33000 Bordeaux, France
- Graduate Program in Neural and Behavioral Science, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Luigi Bellocchio
- INSERM, U1215 NeuroCentre Magendie, Group Endocannabinoids and Neuroadaptation, University of Bordeaux, 33077 Bordeaux, France
| | - Valerie Lemaire
- CNRS, EPHE, INCIA, UMR 5287, Univ. Bordeaux, 33000 Bordeaux, France
| | | | - Bruno Bontempi
- CNRS, EPHE, INCIA, UMR 5287, Univ. Bordeaux, 33000 Bordeaux, France
| | | |
Collapse
|
16
|
Jeste DV, Malaspina D, Bagot K, Barch DM, Cole S, Dickerson F, Dilmore A, Ford CL, Karcher NR, Luby J, Rajji T, Pinto-Tomas AA, Young LJ. Review of Major Social Determinants of Health in Schizophrenia-Spectrum Psychotic Disorders: III. Biology. Schizophr Bull 2023; 49:867-880. [PMID: 37023360 PMCID: PMC10318888 DOI: 10.1093/schbul/sbad031] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
BACKGROUND Social determinants of health (SDoHs) are nonmedical factors that significantly impact health and longevity. We found no published reviews on the biology of SDoHs in schizophrenia-spectrum psychotic disorders (SSPD). STUDY DESIGN We present an overview of pathophysiological mechanisms and neurobiological processes plausibly involved in the effects of major SDoHs on clinical outcomes in SSPD. STUDY RESULTS This review of the biology of SDoHs focuses on early-life adversities, poverty, social disconnection, discrimination including racism, migration, disadvantaged neighborhoods, and food insecurity. These factors interact with psychological and biological factors to increase the risk and worsen the course and prognosis of schizophrenia. Published studies on the topic are limited by cross-sectional design, variable clinical and biomarker assessments, heterogeneous methods, and a lack of control for confounding variables. Drawing on preclinical and clinical studies, we propose a biological framework to consider the likely pathogenesis. Putative systemic pathophysiological processes include epigenetics, allostatic load, accelerated aging with inflammation (inflammaging), and the microbiome. These processes affect neural structures, brain function, neurochemistry, and neuroplasticity, impacting the development of psychosis, quality of life, cognitive impairment, physical comorbidities, and premature mortality. Our model provides a framework for research that could lead to developing specific strategies for prevention and treatment of the risk factors and biological processes, thereby improving the quality of life and increasing the longevity of people with SSPD. CONCLUSIONS Biology of SDoHs in SSPD is an exciting area of research that points to innovative multidisciplinary team science for improving the course and prognosis of these serious psychiatric disorders.
Collapse
Affiliation(s)
- Dilip V Jeste
- Department of Psychiatry, University of California, San Diego (Retired), CA, USA
| | - Dolores Malaspina
- Departments of Psychiatry, Neuroscience and Genetics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kara Bagot
- Department of Psychiatry, Addiction Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deanna M Barch
- Departments of Psychological and Brain Sciences, Psychiatry, and Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Steve Cole
- Departments of Psychiatry and Biobehavioral Sciences, and Medicine, University of California, Los Angeles, CA, USA
| | - Faith Dickerson
- Department of Psychology, Sheppard Pratt, Baltimore, MD, USA
| | - Amanda Dilmore
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Charles L Ford
- Center for Translational Social Neuroscience, Department of Psychiatry, Emory University, Atlanta, GA, USA
| | - Nicole R Karcher
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Joan Luby
- Department of Psychiatry (Child), Washington University in St. Louis, St. Louis, MO, USA
| | - Tarek Rajji
- Adult Neurodevelopment and Geriatric Psychiatry Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Adrián A Pinto-Tomas
- Biochemistry Department, School of Medicine, Universidad de Costa Rica, San José, Costa Rica
| | - Larry J Young
- Center for Translational Social Neuroscience, Department of Psychiatry, Emory University, Atlanta, GA, USA
| |
Collapse
|
17
|
Dothel G, Barbaro MR, Di Vito A, Ravegnini G, Gorini F, Monesmith S, Coschina E, Benuzzi E, Fuschi D, Palombo M, Bonomini F, Morroni F, Hrelia P, Barbara G, Angelini S. New insights into irritable bowel syndrome pathophysiological mechanisms: contribution of epigenetics. J Gastroenterol 2023; 58:605-621. [PMID: 37160449 PMCID: PMC10307698 DOI: 10.1007/s00535-023-01997-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/19/2023] [Indexed: 05/11/2023]
Abstract
Irritable bowel syndrome (IBS) is a complex multifactorial condition including alterations of the gut-brain axis, intestinal permeability, mucosal neuro-immune interactions, and microbiota imbalance. Recent advances proposed epigenetic factors as possible regulators of several mechanisms involved in IBS pathophysiology. These epigenetic factors include biomolecular mechanisms inducing chromosome-related and heritable changes in gene expression regardless of DNA coding sequence. Accordingly, altered gut microbiota may increase the production of metabolites such as sodium butyrate, a prominent inhibitor of histone deacetylases. Patients with IBS showed an increased amount of butyrate-producing microbial phila as well as an altered profile of methylated genes and micro-RNAs (miRNAs). Importantly, gene acetylation as well as specific miRNA profiles are involved in different IBS mechanisms and may be applied for future diagnostic purposes, especially to detect increased gut permeability and visceromotor dysfunctions. In this review, we summarize current knowledge of the role of epigenetics in IBS pathophysiology.
Collapse
Affiliation(s)
- Giovanni Dothel
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Connect By Circular Lab SRL, Madrid, Spain
| | | | - Aldo Di Vito
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Francesca Gorini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Sarah Monesmith
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Emma Coschina
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Eva Benuzzi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Daniele Fuschi
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Marta Palombo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Bonomini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Fabiana Morroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| | - Giovanni Barbara
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Inter-Departmental Center for Health Sciences & Technologies, CIRI-SDV, University of Bologna, Bologna, Italy
| |
Collapse
|
18
|
Mposhi A, Turner JD. How can early life adversity still exert an effect decades later? A question of timing, tissues and mechanisms. Front Immunol 2023; 14:1215544. [PMID: 37457711 PMCID: PMC10348484 DOI: 10.3389/fimmu.2023.1215544] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023] Open
Abstract
Exposure to any number of stressors during the first 1000 days from conception to age 2 years is important in shaping an individual's life trajectory of health and disease. Despite the expanding range of stressors as well as later-life phenotypes and outcomes, the underlying molecular mechanisms remain unclear. Our previous data strongly suggests that early-life exposure to a stressor reduces the capacity of the immune system to generate subsequent generations of naïve cells, while others have shown that, early life stress impairs the capacity of neuronal stem cells to proliferate as they age. This leads us to the "stem cell hypothesis" whereby exposure to adversity during a sensitive period acts through a common mechanism in all the cell types by programming the tissue resident progenitor cells. Furthermore, we review the mechanistic differences observed in fully differentiated cells and suggest that early life adversity (ELA) may alter mitochondria in stem cells. This may consequently alter the destiny of these cells, producing the lifelong "supply" of functionally altered fully differentiated cells.
Collapse
|
19
|
Sun YX, Su YA, Wang Q, Zheng JY, Zhang CC, Wang T, Liu X, Ma YN, Li XX, Zhang XQ, Xie XM, Wang XD, Li JT, Si TM. The causal involvement of the BDNF-TrkB pathway in dentate gyrus in early-life stress-induced cognitive deficits in male mice. Transl Psychiatry 2023; 13:173. [PMID: 37225683 DOI: 10.1038/s41398-023-02476-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 05/03/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
Cognitive dysfunction is a significant, untreated clinical need in patients with psychiatric disorders, for which preclinical studies are needed to understand the underlying mechanisms and to identify potential therapeutic targets. Early-life stress (ELS) leads to long-lasting deficits of hippocampus-dependent learning and memory in adult mice, which may be associated with the hypofunction of the brain-derived neurotrophic factor (BDNF) and its high-affinity receptor, tropomyosin receptor kinase B (TrkB). In this study, we carried out eight experiments using male mice to examine the causal involvement of the BDNF-TrkB pathway in dentate gyrus (DG) and the therapeutic effects of the TrkB agonist (7,8-DHF) in ELS-induced cognitive deficits. Adopting the limited nesting and bedding material paradigm, we first demonstrated that ELS impaired spatial memory, suppressed BDNF expression and neurogenesis in the DG in adult mice. Downregulating BDNF expression (conditional BDNF knockdown) or inhibition of the TrkB receptor (using its antagonist ANA-12) in the DG mimicked the cognitive deficits of ELS. Acute upregulation of BDNF (exogenous human recombinant BDNF microinjection) levels or activation of TrkB receptor (using its agonist, 7,8-DHF) in the DG restored ELS-induced spatial memory loss. Finally, acute and subchronic systemic administration of 7,8-DHF successfully restored spatial memory loss in stressed mice. Subchronic 7,8-DHF treatment also reversed ELS-induced neurogenesis reduction. Our findings highlight BDNF-TrkB system as the molecular target of ELS-induced spatial memory deficits and provide translational evidence for the intervention at this system in the treatment of cognitive deficits in stress-related psychiatric disorders, such as major depressive disorder.
Collapse
Affiliation(s)
- Ya-Xin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Qi Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jia-Ya Zheng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chen-Chen Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ting Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xiao Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Yu-Nu Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xue-Xin Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xian-Qiang Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xiao-Meng Xie
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
- School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
20
|
Tevosian M, Todorov H, Lomazzo E, Bindila L, Ueda N, Bassetti D, Warm D, Kirischuk S, Luhmann HJ, Gerber S, Lutz B. NAPE-PLD deletion in stress-TRAPed neurons results in an anxiogenic phenotype. Transl Psychiatry 2023; 13:152. [PMID: 37149657 PMCID: PMC10164145 DOI: 10.1038/s41398-023-02448-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/08/2023] Open
Abstract
Anandamide (AEA) is an endogenous ligand of the cannabinoid CB1 and CB2 receptors, being a component of the endocannabinoid signaling system, which supports the maintenance or regaining of neural homeostasis upon internal and external challenges. AEA is thought to play a protective role against the development of pathological states after prolonged stress exposure, including depression and generalized anxiety disorder. Here, we used the chronic social defeat (CSD) stress as an ethologically valid model of chronic stress in male mice. We characterized a genetically modified mouse line where AEA signaling was reduced by deletion of the gene encoding the AEA synthesizing enzyme N-acyl-phosphatidylethanolamine-hydrolyzing phospholipase D (NAPE-PLD) specifically in neurons activated at the time of CSD stress. One week after the stress, the phenotype was assessed in behavioral tests and by molecular analyses. We found that NAPE-PLD deficiency in neurons activated during the last three days of CSD stress led to an increased anxiety-like behavior. Investigating the molecular mechanisms underlying this phenotype may suggest three main altered pathways to be affected: (i) desensitization of the negative feedback loop of the hypothalamic-pituitary-adrenal axis, (ii) disinhibition of the amygdala by the prefrontal cortex, and (iii) altered neuroplasticity in the hippocampus and prefrontal cortex.
Collapse
Affiliation(s)
- Margaryta Tevosian
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
| | - Hristo Todorov
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Ermelinda Lomazzo
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Laura Bindila
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Natsuo Ueda
- Department of Biochemistry, Kagawa University School of Medicine, Kagawa, Japan
| | - Davide Bassetti
- Department of Mathematics, Technical University of Kaiserslautern, Kaiserslautern, Germany
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Davide Warm
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Sergei Kirischuk
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany.
| |
Collapse
|
21
|
Sałaciak K, Koszałka A, Lustyk K, Żmudzka E, Jagielska A, Pytka K. Memory impairments in rodent depression models: A link with depression theories. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110774. [PMID: 37088171 DOI: 10.1016/j.pnpbp.2023.110774] [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: 01/13/2023] [Revised: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
More than 80% of depressed patients struggle with learning new tasks, remembering positive events, or concentrating on a single topic. These neurocognitive deficits accompanying depression may be linked to functional and structural changes in the prefrontal cortex and hippocampus. However, their mechanisms are not yet completely understood. We conducted a narrative review of articles regarding animal studies to assess the state of knowledge. First, we argue the contribution of changes in neurotransmitters and hormone levels in the pathomechanism of cognitive dysfunction in animal depression models. Then, we used numerous neuroinflammation studies to explore its possible implication in cognitive decline. Encouragingly, we also observed a positive correlation between increased oxidative stress and a depressive-like state with concomitant memory deficits. Finally, we discuss the undeniable role of neurotrophin deficits in developing cognitive decline in animal models of depression. This review reveals the complexity of depression-related memory impairments and highlights the potential clinical importance of gathered findings for developing more reliable animal models and designing novel antidepressants with procognitive properties.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland
| | - Aleksandra Koszałka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland
| | - Klaudia Lustyk
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland
| | - Elżbieta Żmudzka
- Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College Medyczna, 9 Street, Kraków 30-688, Poland
| | - Angelika Jagielska
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland.
| |
Collapse
|
22
|
Talani G, Biggio F, Gorule AA, Licheri V, Saolini E, Colombo D, Sarigu G, Petrella M, Vedele F, Biggio G, Sanna E. Sex-dependent changes of hippocampal synaptic plasticity and cognitive performance in C57BL/6J mice exposed to neonatal repeated maternal separation. Neuropharmacology 2023; 222:109301. [PMID: 36336069 DOI: 10.1016/j.neuropharm.2022.109301] [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: 03/02/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
The repeated maternal separation (RMS) is a useful experimental model useful in rodents to study the long-term influence of early-life stress on brain neurophysiology. We here investigated the influence of RMS exposure on hippocampal inhibitory and excitatory synaptic transmission, long-term synaptic plasticity and the related potential alterations in learning and memory performance in adult male and female C57Bl/6J mice. Mice were separated daily from their dam for 360 min, from postnatal day 2 (PND2) to PND17, and experiments were performed at PND 60. Patch-clamp recordings in hippocampal CA1 pyramidal neurons revealed a significant enhancement of GABAergic miniature IPSC (mIPSC) frequency, and a decrease in the amplitude of glutamatergic mEPSCs in male mice exposed to RMS. Only a slight but significant reduction in the amplitude of GABAergic mIPSCs was observed in females exposed to RMS compared to the relative controls. A marked increase in long-term depression (LTD) at CA3-CA1 glutamatergic synapses and in the response to the CB1r agonist win55,212 were detected in RMS male, but not female mice. An impaired spatial memory and a reduced preference for novelty was observed in males exposed to RMS but not in females. A single injection of β-ethynyl estradiol at PND2, prevented the changes observed in RMS male mice, suggesting that estrogens may play a protective role early in life against the exposure to stressful conditions. Our findings strengthen the idea of a sex-dependent influence of RMS on long-lasting modifications in synaptic transmission, effects that may be relevant for cognitive performance.
Collapse
Affiliation(s)
- Giuseppe Talani
- CNR Institute of Neuroscience, National Research Council, Monserrato, Italy.
| | - Francesca Biggio
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Ashish Avinash Gorule
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Valentina Licheri
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Eleonora Saolini
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Daniele Colombo
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Gabriele Sarigu
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Michele Petrella
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Francescangelo Vedele
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Giovanni Biggio
- CNR Institute of Neuroscience, National Research Council, Monserrato, Italy; Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| | - Enrico Sanna
- CNR Institute of Neuroscience, National Research Council, Monserrato, Italy; Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, Italy
| |
Collapse
|
23
|
Pelosof R, Santos LAD, Farhat LC, Gattaz WF, Talib L, Brunoni AR. BDNF blood levels after electroconvulsive therapy in patients with mood disorders: An updated systematic review and meta-analysis. World J Biol Psychiatry 2023; 24:24-33. [PMID: 35332840 DOI: 10.1080/15622975.2022.2058083] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVES Studies have suggested Brain-Derived Neurotrophic Factors (BDNF) increase after electroconvulsive therapy (ECT) although they were methodologically limited and enrolled small sample sizes. We aimed at updating a systematic review and meta-analysis to explore BDNF changes after ECT for the treatment of depression. METHODS PubMed, PsycInfo, Embase and Global health were searched (March, 2021). Clinical trials that measured BDNF in the blood before and after ECT in adults (≥ 18 years old) with depression (major depressive disorder or bipolar disorder) were eligible. Data were pooled through random-effects meta-analyses. RESULTS Twenty-eight studies involving 778 participants were included. Meta-analysis showed a significant increase in BDNF levels after ECT (Hedges' g = 0.28; 95% CI: 0.10, 0.46) while there was evidence of significant heterogeneity (I2 = 67.64%) but not publication bias/small-study effect. Subgroup analyses and meta-regressions were underpowered to detect significant differences. Meta-analysis of depression severity scores demonstrated a considerable larger treatment effect in reducing depressive symptoms after ECT (Hedge's g = -3.72 95% CI: -4.23, -3.21). CONCLUSION This updated review showed that BDNF blood levels increased after ECT treatment. However, there was still evidence of substantial heterogeneity and there were limited sample sizes to investigate factors driving the variability of effects across studies. Importantly, the increase in BDNF levels was substantially smaller than the observed in depressive symptomatology, which could be indicative that the former was independent than the latter. Additional studies with larger sample sizes are currently required.
Collapse
Affiliation(s)
- Rebeca Pelosof
- Department of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Leonardo A Dos Santos
- Department of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Luis C Farhat
- Department of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Wagner F Gattaz
- Department of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Service of Interdisciplinary Neuromodulation, Department of Psychiatry, Laboratory of Neurosciences (LIM-27), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Leda Talib
- Department of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Service of Interdisciplinary Neuromodulation, Department of Psychiatry, Laboratory of Neurosciences (LIM-27), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - André R Brunoni
- Department of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Service of Interdisciplinary Neuromodulation, Department of Psychiatry, Laboratory of Neurosciences (LIM-27), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Interdisciplinary Center for Applied Neuromodulation University Hospital, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
24
|
Moon AL, Clifton NE, Wellard N, Thomas KL, Hall J, Brydges NM. Social interaction following prepubertal stress alters prefrontal gene expression associated with cell signalling and oligodendrocytes. Transl Psychiatry 2022; 12:516. [PMID: 36526621 PMCID: PMC9758144 DOI: 10.1038/s41398-022-02280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Early-life adversity is associated with an increased risk of psychopathology, including mood disorders, later in life. Early-life stress affects several physiological systems, however, the exact mechanisms underlying pathological risk are not fully understood. This knowledge is crucial in developing appropriate therapeutic interventions. The prepubertal period is documented as a key developmental period for the maturation of the prefrontal cortex (PFC), a brain region involved in higher cognitive functions, including social function. In this study, we performed RNA sequencing on the PFC of adult rats who had experienced prepubertal stress (PPS) and controls to investigate the genome-wide consequences of this stress. PPS alters social behaviour in adulthood, therefore we also performed RNA sequencing on PPS and control rats following a social interaction test to determine social activity-dependent gene changes. At a baseline state (1 week following a social interaction test), no genes were differentially expressed in the PPS group. However, 1603 genes were differentially expressed in PPS rats compared to controls following a social interaction. These genes were enriched in biological pathways associated with cell signalling and axon myelination dynamics. Cell enrichment analysis showed these genes were associated with oligodendrocytes, and a comparison with an existing early-life stress sequencing dataset showed that pathways linked to oligodendrocyte morphology are impacted in a range of models of early-life stress in rodents. In conclusion, we identify pathways, including those involved in axon myelination, that are differentially activated in the adult in response to social stimulation following PPS. These differential responses may contribute to vulnerability to psychiatric pathology.
Collapse
Affiliation(s)
- Anna L Moon
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Nicholas E Clifton
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.,MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Natalie Wellard
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Kerrie L Thomas
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.,School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.,MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Nichola M Brydges
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
25
|
Kidd T, Dferevine SL, Walker SC. Affective Touch and Regulation of Stress Responses. Health Psychol Rev 2022; 17:60-77. [PMID: 36346350 DOI: 10.1080/17437199.2022.2143854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Much has been documented on the association between stress and health. Both direct and indirect pathways have been identified and explored extensively, helping us understand trajectories from healthy individuals to reductions in well-being, and development of preclinical and disease states. Some of these pathways are well established within the field; physiology, affect regulation, and social relationships. The purpose of this review is to push beyond what is known separately about these pathways and provide a means to integrate them using one common mechanism. We propose that social touch, specifically affective touch, may be the missing active ingredient fundamental to our understanding of how close relationships contribute to stress and health. We provide empirical evidence detailing how affective touch is fundamental to the development of our stress systems, critical to the development of attachment bonds and subsequent social relationships across the life course. We will also explore how we can use this in applied contexts and incorporate it into existing interventions.
Collapse
Affiliation(s)
- Tara Kidd
- School of Psychology, Liverpool John Moore University, Liverpool, U.K
| | | | | |
Collapse
|
26
|
Alizadeh-Ezdini Z, Vatanparast J. Differential impact of two paradigms of early-life adversity on behavioural responses to social defeat in young adult rats and morphology of CA3 pyramidal neurons. Behav Brain Res 2022; 435:114048. [PMID: 35952779 DOI: 10.1016/j.bbr.2022.114048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/20/2022] [Accepted: 08/06/2022] [Indexed: 01/06/2023]
Abstract
Early life stress (ELS) is an important factor in programing the brain for future response to stress, and resilience or vulnerability to stress-induced emotional disorders. The hippocampal formation, with essential roles in both regulating the stress circuitry and emotionality, contributes to this adaptive programing. Here, we examined the effects of early handling (EH) and maternal deprivation (MD) as mild and intense postnatal stressors, respectively, on the behavioural responses to social defeat stress in young adulthood. We also evaluated the interaction of mild and intense ELS with later social defeat (SD) stress on the morphology and dendritic spine density of Golgi-cox-stained CA3 hippocampal neurons. SD stress in adult rats, as expected, increased anxiety and depressive-like behaviours in the open field, elevated plus-maze and forced swimming test. These effects were associated with reduction of dendritic spines and soma size of CA3 neurons. Both behavioural and structural alterations were significantly ameliorated in socially defeated rats that experienced early handling (EH-SD). Basal dendrites of CA3 neurons in EH-SD rats also showed longer dendrites and more intersections with Sholl circles in the distal portion, compared to both control and SD rats. On the other hand, in socially defeated rats with maternal deprivation experience (MD-SD) the stress-induced behavioural and structural alterations were generally intensified compared to SD rats. In MD-SD rats, apical dendrites of CA3 neurons demonstrated remarkable retraction; an effect that was not detected in SD rats. The reduction of dendritic spines density on the apical dendrites of CA3 neurons was also more pronounced in MD-SD rats compared to SD rats. Dendritic arbors and spines comprise the major neuronal substrate for the circuit connectivity, and cell region-specific alterations of dendrites and spines in CA3 neurons reveal plausible mechanisms that can underlie the impact of different ELSs on risk for affective disorders in response to social stress in adulthood.
Collapse
Affiliation(s)
| | - Jafar Vatanparast
- Department of Biology, School of Science, Shiraz University, Shiraz, Iran.
| |
Collapse
|
27
|
Gimsa U, Brückmann R, Tuchscherer A, Tuchscherer M, Kanitz E. Early-life maternal deprivation affects the mother-offspring relationship in domestic pigs, as well as the neuroendocrine development and coping behavior of piglets. Front Behav Neurosci 2022; 16:980350. [PMID: 36275850 PMCID: PMC9582528 DOI: 10.3389/fnbeh.2022.980350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Early-life adversity may have programming effects on the psychological and physiological development of offspring. Domestic pigs (Sus scrofa) are an excellent model species for studying these effects because of their many physiological similarities to humans. Piglets from 10 sows were subjected to daily 2-h maternal deprivation on postnatal days (PND) 2–15 alone (DA) or in a group of littermates (DG). Control piglets (C) from 10 sows stayed with their mothers. Mother-offspring interaction, milk oxytocin, and cortisol were analyzed. An open-field/novel-object (OF/NO) test was performed with piglets on PNDs 16 and 40. Plasma cortisol and immune parameters were determined on PND 5 and 16. Two piglets from each group and sow were sacrificed on PND 20 and stress-related gene expression in the limbic system and prefrontal cortex (PFC), as well as splenic lymphocyte proliferative abilities, were examined. The milk cortisol of sows increased during the first separation of mother and offspring on the second day of lactation, whereas milk oxytocin did not change. The increase in cortisol by the OF/NO test on PND 16 was greater in C piglets than in DA and DG ones. DA piglets showed less agitated behavior than DG and C piglets in the OF/NO test at PND 16, but appeared more fearful. On PND 40, DA piglets showed more arousal than DG and C piglets in the OF/NO test. Neither plasma IgA nor N/L ratios in blood nor mitogen-induced proliferation of spleen lymphocytes were affected by deprivation. We found a higher mRNA expression of CRHR1 in the hypothalamus and a higher expression of MR in the hippocampus in DA piglets than in DG ones. The expression of GR, MR, and CRHR1 genes in the PFC was reduced by maternal deprivation, however, the expression of arginine vasopressin and oxytocin receptors was not affected. Repeated maternal deprivation induces sustained effects on stress reactivity and behavior of domestic piglets. Some of these effects were buffered by the presence of littermates. In addition, we found sex-specific differences in behavior and gene expression.
Collapse
Affiliation(s)
- Ulrike Gimsa
- Psychophysiology Group, Institute of Behavioural Physiology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- *Correspondence: Ulrike Gimsa,
| | - Roberto Brückmann
- Psychophysiology Group, Institute of Behavioural Physiology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- EUROIMMUN Medizinische Labordiagnostika AG, Lübeck, Germany
| | - Armin Tuchscherer
- Service Group Statistical Consulting, Institute of Genetics and Biometry, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Margret Tuchscherer
- Psychophysiology Group, Institute of Behavioural Physiology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Ellen Kanitz
- Psychophysiology Group, Institute of Behavioural Physiology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
28
|
Rosenfield PJ, Jiang D, Pauselli L. Childhood adversity and psychotic disorders: Epidemiological evidence, theoretical models and clinical considerations. Schizophr Res 2022; 247:55-66. [PMID: 34210561 DOI: 10.1016/j.schres.2021.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 02/08/2023]
Abstract
While genetic factors play a critical role in the risk for schizophrenia and other psychotic disorders, increasing evidence points to the role of childhood adversity as one of several environmental factors that can significantly impact the development, manifestations and outcome of these disorders. This paper reviews the epidemiological evidence linking childhood adversity and psychotic disorders and explores various theoretical models that seek to explain the connection. We discuss neurobiological parallels between the impact of childhood trauma and psychosis on the brain and then explore the impact of childhood adversity on different domains of clinical presentation. Finally, implications for prevention and treatment are considered, both on individual and structural levels.
Collapse
Affiliation(s)
- Paul J Rosenfield
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States of America.
| | - David Jiang
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States of America.
| | - Luca Pauselli
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States of America.
| |
Collapse
|
29
|
Loureirin C and Xanthoceraside Prevent Abnormal Behaviors Associated with Downregulation of Brain Derived Neurotrophic Factor and AKT/mTOR/CREB Signaling in the Prefrontal Cortex Induced by Chronic Corticosterone Exposure in Mice. Neurochem Res 2022; 47:2865-2879. [DOI: 10.1007/s11064-022-03694-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/27/2022] [Accepted: 07/14/2022] [Indexed: 11/27/2022]
|
30
|
Thalamocortical bistable switch as a theoretical model of fibromyalgia pathogenesis inferred from a literature survey. J Comput Neurosci 2022; 50:471-484. [PMID: 35816263 PMCID: PMC9666334 DOI: 10.1007/s10827-022-00826-8] [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: 09/07/2021] [Revised: 05/17/2022] [Accepted: 06/22/2022] [Indexed: 11/25/2022]
Abstract
Fibromyalgia (FM) is an unsolved central pain processing disturbance. We aim to provide a unifying model for FM pathogenesis based on a loop network involving thalamocortical regions, i.e., the ventroposterior lateral thalamus (VPL), the somatosensory cortex (SC), and the thalamic reticular nucleus (TRN). The dynamics of the loop have been described by three differential equations having neuron mean firing rates as variables and containing Hill functions to model mutual interactions among the loop elements. A computational analysis conducted with MATLAB has shown a transition from monostability to bistability of the loop behavior for a weakening of GABAergic transmission between TRN and VPL. This involves the appearance of a high-firing-rate steady state, which becomes dominant and is assumed to represent pathogenic pain processing giving rise to chronic pain. Our model is consistent with a bulk of literature evidence, such as neuroimaging and pharmacological data collected on FM patients, and with correlations between FM and immunoendocrine conditions, such as stress, perimenopause, chronic inflammation, obesity, and chronic dizziness. The model suggests that critical targets for FM treatment are to be found among immunoendocrine pathways leading to GABA/glutamate imbalance having an impact on the thalamocortical system.
Collapse
|
31
|
Kent M, Kovalev D, Hart B, Leserve D, Handford G, Vavra D, Lambert K. The emotional impact of disrupted environmental contexts: Enrichment loss and coping profiles influence stress response recovery in Long-Evans rats. J Neuroendocrinol 2022; 34:e13179. [PMID: 35866213 PMCID: PMC9540572 DOI: 10.1111/jne.13179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/20/2022] [Accepted: 06/07/2022] [Indexed: 11/30/2022]
Abstract
With increasing rates of anxiety and mood disorders across the world, there is an unprecedented need for preclinical animal models to generate translational results for humans experiencing disruptive emotional symptoms. Considering that life events resulting in a perception of loss are correlated with depressive symptoms, the enrichment-loss rodent model offers promise as a translational model for stress-initiated psychiatric disorders. Additionally, predisposed temperament characteristics such as coping styles have been found to influence an individual's stress response. Accordingly, male rats were profiled as either consistent or flexible copers and assigned to one of three environments: standard laboratory housing, enriched environment, or enriched environment exposure followed by downsizing to standard laboratory cages (i.e., enrichment-loss group). Throughout the study, several behaviors were assessed to determine stress, social, and reward-processing responses. To assess recovery of the stress response, fecal samples were collected following the swim stress in 3-h increments to determine the recovery trajectory of corticosterone (CORT) and dehydroepiandrosterone (DHEA) metabolite levels. Upon death, neural markers of neuroplasticity including doublecortin, glial fibrillary acidic factor, and brain-derived neurotrophic factor were assessed via immunohistochemistry. Results indicated the flexible coping animals in the continuous enriched group had higher DHEA/CORT ratios (consistent with adaptive responses in past research); furthermore, the enrichment-loss animals exhibited a blunted CORT response throughout the assessments and enriched flexible copers had faster CORT recovery rates than consistent copers. Standard housed animals exhibited less exploratory behavior in the open field task and continuous enriched, flexible rats consumed more food rewards than the other groups. No differences in neuroplasticity neural markers were observed. In sum, the results of the present study support past research indicating the disruptive consequences of enrichment-loss, providing evidence that the model represents a valuable approach for the investigation of neurobiological mechanisms contributing to interindividual variability in responses to changing experiential landscapes.
Collapse
Affiliation(s)
- Molly Kent
- Department of BiologyVirginia Military InstituteLexingtonVAUSA
| | - Dmitry Kovalev
- Department of PsychologyUniversity of RichmondRichmondVAUSA
| | - Benjamin Hart
- Department of PsychologyUniversity of RichmondRichmondVAUSA
| | | | | | - Dylan Vavra
- Department of PsychologyUniversity of RichmondRichmondVAUSA
| | - Kelly Lambert
- Department of PsychologyUniversity of RichmondRichmondVAUSA
| |
Collapse
|
32
|
Early Life Stress Alters Expression of Glucocorticoid Stress Response Genes and Trophic Factor Transcripts in the Rodent Basal Ganglia. Int J Mol Sci 2022; 23:ijms23105333. [PMID: 35628144 PMCID: PMC9141219 DOI: 10.3390/ijms23105333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 01/06/2023] Open
Abstract
Early life stress shapes the developing brain and increases risk for psychotic disorders. Yet, it is not fully understood how early life stress impacts brain regions in dopaminergic pathways whose dysfunction can contribute to psychosis. Therefore, we investigated gene expression following early life stress in adult brain regions containing dopamine neuron cell bodies (substantia nigra, ventral tegmental area (VTA)) and terminals (dorsal/ventral striatum). Sprague-Dawley rats (14F, 10M) were separated from their mothers from postnatal days (PND) 2-14 for 3 h/day to induce stress, while control rats (12F, 10M) were separated for 15 min/day over the same period. In adulthood (PND98), brain regions were dissected, RNA was isolated and five glucocorticoid signalling-related and six brain-derived neurotrophic factor (Bdnf) mRNAs were assayed by qPCR in four brain regions. In the VTA, levels of glucocorticoid signalling-related transcripts differed in maternally separated rodents compared to controls, with the Fkbp5 transcript significantly lower and Ptges3 transcript significantly higher in stressed offspring. In the VTA and substantia nigra, maternally separated rodents had significantly higher Bdnf IIA and III mRNA levels than controls. By contrast, in the ventral striatum, maternally separated rodents had significantly lower expression of Bdnf I, IIA, IIC, IV and VI transcripts. Sex differences in Nr3c1, Bag1 and Fkbp5 expression in the VTA and substantia nigra were also detected. Our results suggest that early life stress has long-lasting impacts on brain regions involved in dopamine neurotransmission, changing the trophic environment and potentially altering responsiveness to subsequent stressful events in a sex-specific pattern.
Collapse
|
33
|
Singh P, Anjum S, Srivastava RK, Tsutsui K, Krishna A. Central and peripheral neuropeptide RFRP-3: A bridge linking reproduction, nutrition, and stress response. Front Neuroendocrinol 2022; 65:100979. [PMID: 35122778 DOI: 10.1016/j.yfrne.2022.100979] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/30/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023]
Abstract
This article is an amalgamation of the current status of RFRP-3 (GnIH) in reproduction and its association with the nutrition and stress-mediated changes in the reproductive activities. GnIH has been demonstrated in the hypothalamus of all the vertebrates studied so far and is a well-known inhibitor of GnRH mediated reproduction. The RFRP-3 neurons interact with the other hypothalamic neurons and the hormonal signals from peripheral organs for coordinating the nutritional, stress, and environmental associated changes to regulate reproduction. RFRP-3 has also been shown to regulate puberty, reproductive cyclicity and senescence depending upon the nutritional status. A favourable nutritional status and the environmental cues which are permissive for the successful breeding and pregnancy outcome keep RFRP-3 level low, whereas unfavourable nutritional status and stressful conditions increase the expression of RFRP-3 which impairs the reproduction. Still our knowledge about RFRP-3 is incomplete regarding its therapeutic application for nutritional or stress-related reproductive disorders.
Collapse
Affiliation(s)
- Padmasana Singh
- Department of Zoology, Indira Gandhi National Tribal University, Amarkantak, Anuppur 484886, MP, India
| | - Shabana Anjum
- Department of Chemical Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Raj Kamal Srivastava
- Department of Zoology, Indira Gandhi National Tribal University, Amarkantak, Anuppur 484886, MP, India
| | - Kazuyoshi Tsutsui
- Department of Biology and Center for Medical Life Science, Waseda University, Kagamiyama 1-7-1, Higashi-Hiroshima University 739-8521, Japan
| | - Amitabh Krishna
- Department of Zoology, Banaras Hindu University, Varanasi 221005, UP, India.
| |
Collapse
|
34
|
Chaudhari PR, Singla A, Vaidya VA. Early Adversity and Accelerated Brain Aging: A Mini-Review. Front Mol Neurosci 2022; 15:822917. [PMID: 35392273 PMCID: PMC8980717 DOI: 10.3389/fnmol.2022.822917] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Early adversity is an important risk factor that influences brain aging. Diverse animal models of early adversity, including gestational stress and postnatal paradigms disrupting dam-pup interactions evoke not only persistent neuroendocrine dysfunction and anxio-depressive behaviors, but also perturb the trajectory of healthy brain aging. The process of brain aging is thought to involve hallmark features such as mitochondrial dysfunction and oxidative stress, evoking impairments in neuronal bioenergetics. Furthermore, brain aging is associated with disrupted proteostasis, progressively defective epigenetic and DNA repair mechanisms, the build-up of neuroinflammatory states, thus cumulatively driving cellular senescence, neuronal and cognitive decline. Early adversity is hypothesized to evoke an “allostatic load” via an influence on several of the key physiological processes that define the trajectory of healthy brain aging. In this review we discuss the evidence that animal models of early adversity impinge on fundamental mechanisms of brain aging, setting up a substratum that can accelerate and compromise the time-line and nature of brain aging, and increase risk for aging-associated neuropathologies.
Collapse
|
35
|
Sampedro‐Piquero P, Moreno‐Fernández RD, Begega A, López M, Santín LJ. Long-term consequences of alcohol use in early adolescent mice: Focus on neuroadaptations in GR, CRF and BDNF. Addict Biol 2022; 27:e13158. [PMID: 35229955 DOI: 10.1111/adb.13158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/11/2022] [Accepted: 01/26/2022] [Indexed: 01/04/2023]
Abstract
Our aim was to assess the cognitive and emotional state, as well as related-changes in the glucocorticoid receptor (GR), the corticotropin-releasing factor (CRF) and the brain-derived neurotrophic factor (BDNF) expression of adolescent C57BL/6J male mice after a 5-week two-bottle choice protocol (postnatal day [pd]21 to pd52). Additionally, we wanted to analyse whether the behavioural and neurobiological effects observed in late adolescence (pd62) lasted until adulthood (pd84). Behavioural testing revealed that alcohol during early adolescence increased anxiety-like and compulsive-related behaviours, which was maintained in adulthood. Concerning cognition, working memory was only altered in late adolescent mice, whereas object location test performance was impaired in both ages. In contrast, novel object recognition remained unaltered. Immunohistochemical analysis showed that alcohol during adolescence diminished BDNF+ cells in the cingulate cortex, the hippocampal CA1 layer and the central amygdala. Regarding hypothalamic-pituitary-adrenal axis (HPA) functioning, alcohol abuse increased the GR and CRF expression in the hypothalamic paraventricular nucleus and the central amygdala. Besides this, GR density was also higher in the prelimbic cortex and the basolateral amygdala, regardless of the animals' age. Our findings suggest that adolescent alcohol exposure led to long-term behavioural alterations, along with changes in BDNF, GR and CRF expression in limbic brain areas involved in stress response, emotional regulation and cognition.
Collapse
Affiliation(s)
- Patricia Sampedro‐Piquero
- Departamento de Psicología Biológica y de la Salud, Facultad de Psicología Universidad Autónoma de Madrid Madrid Spain
| | | | - Azucena Begega
- Departamento de Psicología, Facultad de Psicología Universidad de Oviedo Oviedo Spain
| | - Matías López
- Departamento de Psicología, Facultad de Psicología Universidad de Oviedo Oviedo Spain
| | - Luis J. Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología Universidad de Málaga Málaga Spain
- Neuroimmunology and NeuroInflammation Department Instituto de Investigación Biomédica de Málaga‐IBIMA Málaga Spain
| |
Collapse
|
36
|
Kim S, Gacek SA, Mocchi MM, Redei EE. Sex-Specific Behavioral Response to Early Adolescent Stress in the Genetically More Stress-Reactive Wistar Kyoto More Immobile, and Its Nearly Isogenic Wistar Kyoto Less Immobile Control Strain. Front Behav Neurosci 2022; 15:779036. [PMID: 34970127 PMCID: PMC8713037 DOI: 10.3389/fnbeh.2021.779036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
Genetic predisposition and environmental stress are known etiologies of stress-related psychiatric disorders. Environmental stress during adolescence is assumed to be particularly detrimental for adult affective behaviors. To investigate how genetic stress-reactivity differences modify the effects of stress during adolescence on adult affective behaviors we employed two inbred strains with differing stress reactivity. The Wistar Kyoto More Immobile (WMI) rat strain show increased stress-reactivity and despair-like behaviors as well as passive coping compared to the nearly isogenic control strain, the Wistar Kyoto Less Immobile (WLI). Males and females of these strains were exposed to contextual fear conditioning (CFC) during early adolescence (EA), between 32 and 34 postnatal days (PND), and were tested for the consequences of this mild EA stress in adulthood. Early adolescent stress significantly decreased anxiety-like behavior, measured in the open field test (OFT) and increased social interaction and recognition in adult males of both strains compared to controls. In contrast, no significant effects of EA stress were observed in adult females in these behaviors. Both males and females of the genetically less stress-reactive WLI strain showed significantly increased immobility in the forced swim test (FST) after EA stress compared to controls. In contrast, immobility was significantly attenuated by EA stress in adult WMI females compared to controls. Transcriptomic changes of the glucocorticoid receptor (Nr3c1, GR) and the brain-derived neurotrophic factor (Bdnf) illuminate primarily strain and stress-dependent changes, respectively, in the prefrontal cortex and hippocampus of adults. These results suggest that contrary to expectations, limited adolescent stress is beneficial to males thru decreasing anxiety and enhancing social behaviors, and to the stress more-reactive WMI females by way of decreasing passive coping.
Collapse
Affiliation(s)
- Sarah Kim
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Stephanie A Gacek
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Madaline M Mocchi
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Eva E Redei
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
37
|
Natale NR, Kent M, Fox N, Vavra D, Lambert K. Neurobiological effects of a probiotic-supplemented diet in chronically stressed male Long-Evans rats: Evidence of enhanced resilience. IBRO Neurosci Rep 2021; 11:207-215. [PMID: 34849506 PMCID: PMC8607205 DOI: 10.1016/j.ibneur.2021.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
Probiotics that regulate the microbiome-gut-brain axis and provide mental health benefits to the host are referred to as psychobiotics. Preclinical studies have demonstrated psychobiotic effects on early life stress-induced anxiety- and depression-related behavior in rodents; however, the specific mechanisms remain ill-defined. In the current study, we investigated the effects of probiotic supplementation on neurobiological responses to chronic stress in adult male Long-Evans rats. Twenty-four rats were randomly assigned to probiotic (PB) or vehicle control (VEH) groups, then to either chronic unpredictable stress (CUS) or no-stress control (CON) conditions within each group (n = 6/subgroup). We hypothesized that PB supplementation would reduce markers of anxiety and enhance emotional resilience, especially in the CUS animals. In the cognitive uncertainty task, a nonsignificant trend was observed indicating that the PB-supplemented animals spent more time oriented toward the food reward than VEH animals. In the open-field task, CUS-PB animals spent more time in the center of the arena than CUS-VEH animals, an effect not observed between the two CON groups. In the swim task, the PB animals, regardless of stress assignment, exhibited increased floating, suggesting a conserved response in a challenging context. Focusing on the endocrine measures, higher dehydroepiandrosterone (DHEA)-to-corticosterone fecal metabolite ratios, a correlate of emotional resilience, were observed in PB animals. Further, PB animals exhibited reduced microglia immunoreactivity in the basolateral amygdala, possibly indicating a neuroprotective effect of PB supplements in this rodent model. These results provide evidence that PB supplementation interacts with stress exposure to influence adaptive responses associated with endocrine, neural, and behavioral indices of anxiety.
Collapse
Affiliation(s)
- Nick R. Natale
- Dept of Psychology, University of Richmond, VA 23173, USA
| | - Molly Kent
- Dept of Biology, Virginia Military Institute, Lexington, VA 24450, USA
| | - Nathan Fox
- Dept of Psychology, University of Richmond, VA 23173, USA
| | - Dylan Vavra
- Dept of Psychology, University of Richmond, VA 23173, USA
| | - Kelly Lambert
- Dept of Psychology, University of Richmond, VA 23173, USA
| |
Collapse
|
38
|
Timmerman BM, Mooney-Leber SM, Brummelte S. The effects of neonatal procedural pain and maternal isolation on hippocampal cell proliferation and reelin concentration in neonatal and adult male and female rats. Dev Psychobiol 2021; 63:e22212. [PMID: 34813104 DOI: 10.1002/dev.22212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 11/11/2022]
Abstract
Preterm births accounted for over 10% of all U.S. live births in 2019 and the rate is rising. Neonatal stressors, especially procedural pain, experienced by preterm infants in the neonatal intensive care unit (NICU) have been associated with neurodevelopmental impairments. Parental care can alleviate stress during stressful or painful procedures; however, infants in the NICU often receive reduced parental care compared with their peers. Animal studies suggest that decreased maternal care similarly impairs neurodevelopment but also influences the effects of neonatal pain. It is important to mimic both stressors in animal models of neonatal stress exposure. In this study, researchers investigated the individual and combined impact of neonatal pain and maternal isolation on reelin protein levels and cellular proliferation in the hippocampal dentate gyrus of 8 days old and adult rats. Exposure to either stressor individually, but not both, increased reelin levels in the dentate gyrus of adult females without significantly altering reelin levels in adult males. However, cell proliferation levels at either age were unaffected by the early-life stressors. These results suggest that each early-life stressor has a unique effect on markers of brain development and more research is needed to further investigate their distinct influences.
Collapse
Affiliation(s)
- Brian M Timmerman
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
| | - Sean M Mooney-Leber
- Department of Psychology, University of Wisconsin-Stevens Points, Stevens Point, Wisconsin, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, Michigan, USA.,Translational Neuroscience Program, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
39
|
Cruciani G, Boccia M, Lingiardi V, Giovanardi G, Zingaretti P, Spitoni GF. An Exploratory Study on Resting-State Functional Connectivity in Individuals with Disorganized Attachment: Evidence for Key Regions in Amygdala and Hippocampus. Brain Sci 2021; 11:brainsci11111539. [PMID: 34827538 PMCID: PMC8615787 DOI: 10.3390/brainsci11111539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 02/02/2023] Open
Abstract
Studies comparing organized (O) and unresolved/disorganized (UD) attachment have consistently shown structural and functional brain abnormalities, although whether and how attachment patterns may affect resting state functional connectivity (RSFC) is still little characterized. Here, we investigated RSFC of temporal and limbic regions of interest for UD attachment. Participants’ attachment was classified via the Adult Attachment Interview, and all participants underwent clinical assessment. Functional magnetic resonance imaging data were collected from 11 UD individuals and seven matched O participants during rest. A seed-to-voxel analysis was performed, including the anterior and the posterior cingulate cortex, the bilateral insula, amygdala and hippocampus as seed regions. No group differences in the clinical scales emerged. Compared to O, the UD group showed lower RSFC between the left amygdala and the left cerebellum (lobules VIII), and lower functional coupling between the right hippocampus and the posterior portion of the right middle temporal gyrus. Moreover, UD participants showed higher RSFC between the right amygdala and the anterior cingulate cortex. Our findings suggest RSFC alterations in regions associated with encoding of salient events, emotion processing, memories retrieval and self-referential processing in UD participants, highlighting the potential role of attachment experiences in shaping brain abnormalities also in non-clinical UD individuals.
Collapse
Affiliation(s)
- Gianluca Cruciani
- Department of Psychology, Ph.D. Program in Behavioral Neuroscience, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
- Correspondence: ; Tel.: +39-(0)6-49917711
| | - Maddalena Boccia
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy;
- Cognitive and Motor Rehabilitation and Neuroimaging Unit, Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00179 Rome, Italy;
| | - Vittorio Lingiardi
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, 00185 Rome, Italy; (V.L.); (G.G.)
| | - Guido Giovanardi
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, 00185 Rome, Italy; (V.L.); (G.G.)
| | - Pietro Zingaretti
- Villa von Siebenthal Neuropsychiatric Clinic and Hospital, Genzano di Roma, 00045 Rome, Italy;
| | - Grazia Fernanda Spitoni
- Cognitive and Motor Rehabilitation and Neuroimaging Unit, Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00179 Rome, Italy;
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, 00185 Rome, Italy; (V.L.); (G.G.)
| |
Collapse
|
40
|
Jiang Z, Zhu Z, Zhao M, Wang W, Li H, Liu D, Pan F. H3K9me2 regulation of BDNF expression in the hippocampus and medial prefrontal cortex is involved in the depressive-like phenotype induced by maternal separation in male rats. Psychopharmacology (Berl) 2021; 238:2801-2813. [PMID: 34328517 DOI: 10.1007/s00213-021-05896-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Early life stress (ELS) induces a depressive-like phenotype and increases the risk of depression. Brain-derived neurotrophic factor (BDNF) has been confirmed to be involved in the pathophysiology of depression. However, the mechanism by which ELS alters the epigenetic regulation of BDNF and changes susceptibility to depression has not been fully clarified. METHODS The present study used maternal separation (MS) and chronic unpredicted mild stress (CUMS) to establish an MS animal model and a depressive animal model. We assessed depressive-like behaviours, including anhedonia, locomotor activity, anxiety-like behaviour, and spatial memory, using the sucrose preference test, the open field test, the elevated plus maze test, and the Morris water maze test. We also investigated BDNF and H3K9me2 expression in the hippocampus and medial prefrontal cortex (mPFC) by immunohistochemistry, western blotting, and qPCR analysis. Additionally, we used Unc0642, a small molecule inhibitor of histone methyltransferase (G9a), as an intervention. RESULTS The results showed that CUMS induced depressive-like behaviours in rats and resulted in increased H3K9me2 expression and decreased BDNF expression in the hippocampus and mPFC. More importantly, adult MS rats experiencing CUMS had more severe depressive behaviours, had higher expression of H3K9me2 in the hippocampus and mPFC, and had lower expression of BDNF in the hippocampus and mPFC. In addition, administration of the G9a inhibitor reversed most of the changes. CONCLUSIONS Our study suggests that ELS changed BDNF and H3K9me2 expression in the rat brain, resulting in a depressive-like phenotype.
Collapse
Affiliation(s)
- Zhijun Jiang
- Department of Medical Psychology and Ethics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Zemeng Zhu
- Department of Medical Psychology and Ethics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Mingyue Zhao
- Department of Medical Psychology and Ethics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Wei Wang
- Department of Medical Psychology and Ethics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Haonan Li
- Department of Medical Psychology and Ethics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Fang Pan
- Department of Medical Psychology and Ethics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
41
|
Neuroplasticity and Multilevel System of Connections Determine the Integrative Role of Nucleus Accumbens in the Brain Reward System. Int J Mol Sci 2021; 22:ijms22189806. [PMID: 34575969 PMCID: PMC8471564 DOI: 10.3390/ijms22189806] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
A growing body of evidence suggests that nucleus accumbens (NAc) plays a significant role not only in the physiological processes associated with reward and satisfaction but also in many diseases of the central nervous system. Summary of the current state of knowledge on the morphological and functional basis of such a diverse function of this structure may be a good starting point for further basic and clinical research. The NAc is a part of the brain reward system (BRS) characterized by multilevel organization, extensive connections, and several neurotransmitter systems. The unique role of NAc in the BRS is a result of: (1) hierarchical connections with the other brain areas, (2) a well-developed morphological and functional plasticity regulating short- and long-term synaptic potentiation and signalling pathways, (3) cooperation among several neurotransmitter systems, and (4) a supportive role of neuroglia involved in both physiological and pathological processes. Understanding the complex function of NAc is possible by combining the results of morphological studies with molecular, genetic, and behavioral data. In this review, we present the current views on the NAc function in physiological conditions, emphasizing the role of its connections, neuroplasticity processes, and neurotransmitter systems.
Collapse
|
42
|
Affiliation(s)
- Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK
| |
Collapse
|
43
|
Yu Z, Bai R, Zhou J, Huang H, Zhao W, Huo X, Yang Y, Luan Z, Zhang B, Sun C, Ma X. Uncarialins J—M from
Uncaria rhynchophylla
and Their Anti‐depression Mechanism in Unpredictable Chronic Mild
Stress‐Induced
Mice
via
Activating
5‐HT
1A
Receptor. CHINESE J CHEM 2021. [DOI: 10.1002/cjoc.202000652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Zhen‐Long Yu
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University Dalian Liaoning 116044 China
| | - Rong Bai
- Department of Pharmacy, Shanghai East Hospital, Tongji University Shanghai 200120 China
| | - Jun‐Jun Zhou
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University Dalian Liaoning 116044 China
| | - Hui‐Lian Huang
- Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang Jiangxi 330103 China
| | - Wen‐Yu Zhao
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University Dalian Liaoning 116044 China
| | - Xiao‐Kui Huo
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University Dalian Liaoning 116044 China
| | - Ya‐Hui Yang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University Dalian Liaoning 116044 China
| | - Zhi‐Lin Luan
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University Dalian Liaoning 116044 China
| | - Bao‐Jing Zhang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University Dalian Liaoning 116044 China
| | - Cheng‐Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University Dalian Liaoning 116044 China
| | - Xiao‐Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, College (Institute) of Integrative Medicine, Dalian Medical University Dalian Liaoning 116044 China
| |
Collapse
|
44
|
The role of pre-pubertal training history on hippocampal neurotrophic factors and glucocorticoid receptor protein levels in adult male rats. Neurosci Lett 2021; 752:135834. [PMID: 33771578 DOI: 10.1016/j.neulet.2021.135834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 11/22/2022]
Abstract
Neurotrophic factors play an integral role in hippocampal plasticity, and interaction with HPA axis components, especially glucocorticoid receptors (GR), can mediate the structural and functional changes. In the present work, we investigated the long-term effects of combined exercise training (CET) and voluntary physical activity in an enriched environment (EE) in the pre-pubertal period on hippocampal neurotrophic factors and GR. For this purpose, a longitudinal study was designed. After three weeks, all rats were kept in the standard cages without any interventions until adulthood. Western blot analysis revealed a significant increase in hippocampal BDNF and VEGF protein levels in both EE and CET groups (P < 0.001), along with an increase in GR protein levels. In addition, EE decreased serum corticosterone levels compared to CET (P < 0.05). Serum insulin-like growth factor-1 (IGF-1) levels did not demonstrate remarkable changes between groups. Training interventions during sensitive developmental periods may produce profound and long-lasting effects on the hippocampus, at least in part by interactive effects of neurotrophic factors cascades and GR.
Collapse
|
45
|
Staurengo-Ferrari L, Green PG, Araldi D, Ferrari LF, Miaskowski C, Levine JD. Sexual dimorphism in the contribution of neuroendocrine stress axes to oxaliplatin-induced painful peripheral neuropathy. Pain 2021; 162:907-918. [PMID: 32947545 PMCID: PMC7886966 DOI: 10.1097/j.pain.0000000000002073] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/24/2020] [Indexed: 01/05/2023]
Abstract
ABSTRACT Although clinical studies support the suggestion that stress is a risk factor for painful chemotherapy-induced peripheral neuropathy (CIPN), there is little scientific validation to support this link. Here, we evaluated the impact of stress on CIPN induced by oxaliplatin, and its underlying mechanisms, in male and female rats. A single dose of oxaliplatin produced mechanical hyperalgesia of similar magnitude in both sexes, still present at similar magnitude in both sexes, on day 28. Adrenalectomy mitigated oxaliplatin-induced hyperalgesia, in both sexes. To confirm the role of neuroendocrine stress axes in CIPN, intrathecal administration of antisense oligodeoxynucleotide targeting β₂-adrenergic receptor mRNA both prevented and reversed oxaliplatin-induced hyperalgesia, only in males. By contrast, glucocorticoid receptor antisense oligodeoxynucleotide prevented and reversed oxaliplatin-induced hyperalgesia in both sexes. Unpredictable sound stress enhanced CIPN, in both sexes. The administration of stress hormones, epinephrine, corticosterone, and their combination, at stress levels, mimicked the effects of sound stress on CIPN, in males. In females, only corticosterone mimicked the effect of sound stress. Also, a risk factor for CIPN, early-life stress, was evaluated by producing both stress-sensitive (produced by neonatal limited bedding) and stress-resilient (produced by neonatal handling) phenotypes in adults. Although neonatal limited bedding significantly enhanced CIPN only in female adults, neonatal handling significantly attenuated CIPN, in both sexes. Our study demonstrates a sexually dimorphic role of the 2 major neuroendocrine stress axes in oxaliplatin-induced neuropathic pain.
Collapse
Affiliation(s)
- Larissa Staurengo-Ferrari
- Departments of Medicine and Oral & Maxillofacial Surgery, Division of Neuroscience, UCSF Pain and Addiction Research Center, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Paul G. Green
- Departments of Preventative & Restorative Dental Sciences and Oral & Maxillofacial Surgery, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Dionéia Araldi
- Departments of Medicine and Oral & Maxillofacial Surgery, Division of Neuroscience, UCSF Pain and Addiction Research Center, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Luiz F. Ferrari
- Departments of Medicine and Oral Surgery, and Division of Neuroscience, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA. Current address: Department of Anesthesiology, University of Utah, 30 N Medical Dr. RM 3C4444, Salt Lake City, UT 84132
| | - Christine Miaskowski
- Departments of Physiological Nursing and Anesthesia, UCSF Pain and Addiction Research Center, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Jon D. Levine
- Departments of Medicine and Oral & Maxillofacial Surgery, Division of Neuroscience, UCSF Pain and Addiction Research Center, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| |
Collapse
|
46
|
The downstream effects of forced exercise training and voluntary physical activity in an enriched environment on hippocampal plasticity in preadolescent rats. Brain Res 2021; 1759:147373. [PMID: 33600831 DOI: 10.1016/j.brainres.2021.147373] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/02/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022]
Abstract
During critical periods of brain development, exercise-induced physical fitness may greatly impact the brain structure and function. Nevertheless, forced and intensive physical activities may display negative effects, particularly in the pre-pubertal period. Preadolescent rats were exposed to an enriched environment and combined exercise training for three consecutive weeks in the present study. There was a large cage with enriching stimuli and voluntary physical activity opportunities as an enriched environment (EE). The combined exercise training (CET) consisted of aerobic and resistance training programs. The protein levels of corticosterone (CORT), glucocorticoid receptors (GRs), insulin-like growth factor-1 (IGF-1), brain-derived neurotrophic factor (BDNF), and vascular endothelial growth factor (VEGF) were assessed using Enzyme-linked immunosorbent assay and western blotting. Cresyl violet staining was also used to evaluate the number of cells in the hippocampus. While GRs levels were significantly increased in both EE and CET groups (P < 0.001), decreased CORT levels were found in enriched rats (P < 0.05). Moreover, elevated BDNF levels were found in the EE (P < 0.01) and CET (P < 0.05) groups. Similarly, VEGF significantly increased in the EE (P < 0.01) and CET (P < 0.05) animals. However, IGF-1 levels were high only in trained rats (P < 0.05). The number of cells also significantly increased in the DG and CA1 region of the hippocampus after each intervention (P < 0.001). These findings clarified that combined exercise training and voluntary physical activity in an enriched environment during the preadolescent period might promote the downstream plasticity effects on the hippocampus.
Collapse
|
47
|
Mancini GF, Marchetta E, Pignani I, Trezza V, Campolongo P. Social Defeat Stress During Early Adolescence Confers Resilience Against a Single Episode of Prolonged Stress in Adult Rats. Cells 2021; 10:360. [PMID: 33572375 PMCID: PMC7916240 DOI: 10.3390/cells10020360] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 01/19/2023] Open
Abstract
Early-life adverse experiences (first hit) lead to coping strategies that may confer resilience or vulnerability to later experienced stressful events (second hit) and the subsequent development of stress-related psychopathologies. Here, we investigated whether exposure to two stressors at different stages in life has long-term effects on emotional and cognitive capabilities, and whether the interaction between the two stressors influences stress resilience. Male rats were subjected to social defeat stress (SDS, first hit) in adolescence and to a single episode of prolonged stress (SPS, second hit) in adulthood. Behavioral outcomes, hippocampal expression of brain-derived neurotrophic factor, and plasma corticosterone levels were tested in adulthood. Rats exposed to both stressors exhibited resilience against the development of stress-induced alterations in emotional behaviors and spatial memory, but vulnerability to cued fear memory dysfunction. Rats subjected to both stressors demonstrated resilience against the SDS-induced alterations in hippocampal brain-derived neurotrophic factor expression and plasma corticosterone levels. SPS alone altered locomotion and spatial memory retention; these effects were absent in SDS-exposed rats later exposed to SPS. Our findings reveal that exposure to social stress during early adolescence influences the ability to cope with a second challenge experienced later in life.
Collapse
Affiliation(s)
- Giulia Federica Mancini
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (G.F.M.); (E.M.); (I.P.)
- Neurobiology of Behavior Laboratory, Santa Lucia Foundation, 00143 Rome, Italy
| | - Enrico Marchetta
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (G.F.M.); (E.M.); (I.P.)
- Neurobiology of Behavior Laboratory, Santa Lucia Foundation, 00143 Rome, Italy
| | - Irene Pignani
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (G.F.M.); (E.M.); (I.P.)
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University Roma Tre, 00146 Rome, Italy;
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (G.F.M.); (E.M.); (I.P.)
- Neurobiology of Behavior Laboratory, Santa Lucia Foundation, 00143 Rome, Italy
| |
Collapse
|
48
|
Babicola L, Ventura R, D'Addario SL, Ielpo D, Andolina D, Di Segni M. Long term effects of early life stress on HPA circuit in rodent models. Mol Cell Endocrinol 2021; 521:111125. [PMID: 33333214 DOI: 10.1016/j.mce.2020.111125] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/23/2020] [Accepted: 12/10/2020] [Indexed: 01/06/2023]
Abstract
Adaptation to environmental challenges represents a critical process for survival, requiring the complex integration of information derived from both external cues and internal signals regarding current conditions and previous experiences. The Hypothalamic-pituitary-adrenal axis plays a central role in this process inducing the activation of a neuroendocrine signaling cascade that affects the delicate balance of activity and cross-talk between areas that are involved in sensorial, emotional, and cognitive processing such as the hippocampus, amygdala, Prefrontal Cortex, Ventral Tegmental Area, and dorsal raphe. Early life stress, especially early critical experiences with caregivers, influences the functional and structural organization of these areas, affects these processes in a long-lasting manner and may result in long-term maladaptive and psychopathological outcomes, depending on the complex interaction between genetic and environmental factors. This review summarizes the results of studies that have modeled this early postnatal stress in rodents during the first 2 postnatal weeks, focusing on the long-term effects on molecular and structural alteration in brain areas involved in Hypothalamic-pituitary-adrenal axis function. Moreover, a brief investigation of epigenetic mechanisms and specific genetic targets mediating the long-term effects of these early environmental manipulations and at the basis of differential neurobiological and behavioral effects during adulthood is provided.
Collapse
Affiliation(s)
- Lucy Babicola
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Rossella Ventura
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy.
| | - Sebastian Luca D'Addario
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy; Behavioral Neuroscience PhD Programme, Sapienza University, Piazzale Aldo Moro 5, 00184, Rome, Italy
| | - Donald Ielpo
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy; Behavioral Neuroscience PhD Programme, Sapienza University, Piazzale Aldo Moro 5, 00184, Rome, Italy
| | - Diego Andolina
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, 00184, Rome, Italy; IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Matteo Di Segni
- IRCCS Fondazione Santa Lucia, Via Del Fosso di Fiorano, 64, 00143, Rome, Italy.
| |
Collapse
|
49
|
Luft C, Levices IP, da Costa MS, de Oliveira JR, Donadio MVF. Effects of running before pregnancy on long-term memory and hippocampal alterations induced by prenatal stress. Neurosci Lett 2021; 746:135659. [PMID: 33482306 DOI: 10.1016/j.neulet.2021.135659] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/05/2021] [Accepted: 01/10/2021] [Indexed: 12/22/2022]
Abstract
Studies have shown that an adverse environment in utero influences fetal growth and development, leading to several neuroendocrine and behavioral changes in adult life. Nevertheless, the mechanisms involved in the long-term benefits of pregestational exercise are still poorly understood. Thus, this study aimed to evaluate the effects of physical exercise before the gestational period on memory behavior and gene expression in the hippocampus of adult mice submitted to prenatal stress. Female Balb/c mice were divided into three groups: control (CON), prenatal restraint stress (PNS), and exercise before the gestational period plus PNS (EX + PNS). When adults, male and female offspring were submitted to the object recognition test followed by the hippocampal evaluation of BDNF exons I and IV mRNA expression, as well as hypothalamic-pituitary-adrenal axis related genes. Pregestational exercise did not prevent the decreased recognition index, as well as GR and CRHR1 gene expression observed in PNS males. Conversely, prenatal stress did not influence female memory behavior. Moreover, exercise attenuated the effects of prenatal stress on female BDNF IV gene expression. The results indicate that pregestational exercise was able to prevent the effects of maternal stress on hippocampal BDNF IV gene expression in females, although no effects were seen on the stress-induced memory impairment in males.
Collapse
Affiliation(s)
- Carolina Luft
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Isadora Perez Levices
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Mariana Severo da Costa
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Márcio Vinícius Fagundes Donadio
- Laboratory of Pediatric Physical Activity, Infant Center, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Laboratory of Cellular Biophysics and Inflammation, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
| |
Collapse
|
50
|
Algamal M, Pearson AJ, Hahn-Townsend C, Burca I, Mullan M, Crawford F, Ojo JO. Repeated unpredictable stress and social isolation induce chronic HPA axis dysfunction and persistent abnormal fear memory. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110035. [PMID: 32682873 DOI: 10.1016/j.pnpbp.2020.110035] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/08/2020] [Accepted: 07/12/2020] [Indexed: 12/19/2022]
Abstract
The lack of progress in the psychopharmacological treatment of stress-related disorders such as PTSD is an ongoing crisis due to its negative socioeconomic implications. Current PTSD pharmacotherapy relies on a few FDA approved medications used primarily for depression which offer only symptomatic relief and show limited efficacy. As the population of PTSD patients is growing, the identification of effective etiology-based treatments for the condition is a high priority. This requires an in-depth understanding of the neurobiological and behavioral outcomes of stress in translationally relevant animal models. In this study, we use neuroendocrine, biochemical and behavioral measures to assess the HPA axis function and fear-memory deficits in a mouse model of chronic stress. The chronic stress procedures involved exposure to 21 days of repeated unpredictable stress (RUS), including predator stress, restraint and foot shock, followed by chronic social isolation. We show that mice exposed to our stress paradigm demonstrate exaggerated fear memory recall and blunted HPA axis functionality at one month after RUS. Our neuroendocrinal testing suggests that the attenuated stress response in our model may be related to an alteration in the adrenal MC2 receptor reactivity. While there was no noticeable change in pituitary negative feedback regulation mechanisms, CRH and phosphorylated Glucocorticoid receptors levels were altered in the hypothalamus. We also show that chronic supplementation with a peripheral glucocorticoid receptor agonist (low-dose dexamethasone) after RUS partially restores a number of stress-related behavioral deficits in the RUS model. This suggests a direct relationship between HPA axis function and behavior in our model. Our findings emphasize the importance of the adrenal receptors as a target for HPA axis dysfunction in stress and fear-related disorders.
Collapse
Affiliation(s)
- Moustafa Algamal
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL 34243, USA; The Open University, Milton Keynes, United Kingdom; James A. Haley Veterans' Hospital, Tampa, FL, United States.
| | - Andrew J Pearson
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL 34243, USA; The Open University, Milton Keynes, United Kingdom; James A. Haley Veterans' Hospital, Tampa, FL, United States.
| | | | - Ioana Burca
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL 34243, USA.
| | - Michael Mullan
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL 34243, USA; The Open University, Milton Keynes, United Kingdom.
| | - Fiona Crawford
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL 34243, USA; The Open University, Milton Keynes, United Kingdom; James A. Haley Veterans' Hospital, Tampa, FL, United States.
| | - Joseph O Ojo
- Roskamp Institute, 2040 Whitfield Ave, Sarasota, FL 34243, USA; The Open University, Milton Keynes, United Kingdom; James A. Haley Veterans' Hospital, Tampa, FL, United States.
| |
Collapse
|