151
|
Neubert da Silva G, Zauer Curi T, Lima Tolouei SE, Tapias Passoni M, Sari Hey GB, Marino Romano R, Martino-Andrade AJ, Dalsenter PR. Effects of diisopentyl phthalate exposure during gestation and lactation on hormone-dependent behaviours and hormone receptor expression in rats. J Neuroendocrinol 2019; 31:e12816. [PMID: 31758603 DOI: 10.1111/jne.12816] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/28/2022]
Abstract
Phthalates are found in different plastic materials, such as packaging, toys and medical devices. Some of these compounds are endocrine disruptors, comprising substances that are able to induce multiple hormonal disturbances and downstream developmental effects, including the disruption of androgen-dependent differentiation of the male reproductive tract and changes in pathways that regulate hormone-dependent behaviours. In a previous study, metabolites of diisopentyl phthalate (DiPeP), a potent anti-androgenic phthalate, were found in the urine of Brazilian pregnant women. Therefore, the present study aimed to evaluate the effects of DiPeP exposure during critical developmental periods on behaviours controlled by sex hormones in rats. Pregnant Wistar rats were treated with DiPeP (1, 10 or 100 mg kg day-1 ) or canola oil by oral gavage between gestational day 10 and post-natal day (PND) 21. Male offspring were tested in a behavioural battery, including the elevated plus maze task, play behaviour, partner preference and sexual behaviour. After the behavioural tests, the hypothalamus and pituitary of these animals were removed on PND 60-65 and PND 145-160 to quantify gene expression for aromatase, androgen receptor (Ar) and oestrogen receptors α (Esr1) and β (Esr2). Male rats exposed to 1 and 10 mg kg day-1 DiPeP displayed no preference for the female stimulus rat in the partner preference test and 1 mg kg day-1 DiPeP rats also showed a significant increase in mount and penetration latencies when mated with receptive females. A decrease in pituitary Esr1 expression was observed in all DiPeP treated groups regardless of age. A reduction in hypothalamic Esr1 expression in rats exposed to 10 mg kg day-1 DiPeP was also observed. No significant changes were found with respect to Ar, Esr2 and aromatase expression in the hypothalamus. These results suggest that DiPeP exposure during critical windows of development in rats may induce changes in behaviours related to mating and the sexual motivation of males.
Collapse
|
152
|
Prefrontal cortex interneurons display dynamic sex-specific stress-induced transcriptomes. Transl Psychiatry 2019; 9:292. [PMID: 31712551 PMCID: PMC6848179 DOI: 10.1038/s41398-019-0642-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022] Open
Abstract
γ-aminobutyric acid (GABA) inhibitory interneurons play a key role in efferent and afferent control of principle neuron activity in the prefrontal cortex (PFC), thereby regulating signal integrity of cognitive and behavioral processes. Recent evidence suggests that specific subtypes of interneurons in the PFC mediate stress-induced depressive-like behaviors. Abnormalities of GABA interneurons, particularly the somatostatin (human, SST; mouse, Sst) subtype, have been reported in postmortem brains of depressed subjects and include sex differences that could explain the increased incidence of depression in women. Here, we analyze the transcriptional profiles and the effects of chronic stress in males vs. females on GABA interneuron subtypes in the PFC. Using Sst- and Parvalbumin-fluorescence tagged reporter mice and fluorescence-activated cell sorting (FACS) combined with RNA sequencing, we identify distinct transcriptome profiles for these interneuron subtypes in the medial PFC. Based on evidence that SST interneurons are altered in depression, we then determined the effects of chronic stress on this interneuron subtype. Chronic stress causes significant dysregulation of several key pathways, including sex-specific differences in the Sst interneuron profiles. The transcriptional pathways altered by chronic stress in males overlap with enriched pathways in non-stressed females. These changes occurred predominantly in decreased expression of elongation initiation factor 2 (EIF2) signaling, suggesting that dysfunction of the translational machinery of SST interneurons could be critical to the development of depressive-like behaviors in males. In addition, SST interneurons from females exposed to chronic stress show dysregulation of different, growth factor signaling pathways.
Collapse
|
153
|
Kokras N, Hodes GE, Bangasser DA, Dalla C. Sex differences in the hypothalamic-pituitary-adrenal axis: An obstacle to antidepressant drug development? Br J Pharmacol 2019; 176:4090-4106. [PMID: 31093959 PMCID: PMC6877794 DOI: 10.1111/bph.14710] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 12/30/2022] Open
Abstract
Hypothalamic-pituitary-adrenal (HPA) axis dysfunction has long been implicated in the pathophysiology of depression, and HPA axis-based compounds have served as potential new therapeutic targets, but with no success. This review details sex differences from animal and human studies in the function of HPA axis elements (glucocorticoids, corticotropin releasing factor, and vasopressin) and related compounds tested as candidate antidepressants. We propose that sex differences contribute to the failure of novel HPA axis-based drugs in clinical trials. Compounds studied preclinically in males were tested in clinical trials that recruited more, if not exclusively, women, and did not control, but rather adjusted, for potential sex differences. Indeed, clinical trials of antidepressants are usually not stratified by sex or other important factors, although preclinical and epidemiological data support such stratification. In conclusion, we suggest that clinical testing of HPA axis-related compounds creates an opportunity for targeted, personalized antidepressant treatments based on sex. LINKED ARTICLES: This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Nikolaos Kokras
- Department of PharmacologyNational and Kapodistrian University of AthensAthensGreece
- First Department of Psychiatry, Eginition HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Georgia E. Hodes
- School of NeuroscienceVirginia Polytechnic Institute and State UniversityBlacksburgVirginia
| | | | - Christina Dalla
- Department of PharmacologyNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
154
|
An alternative theory for hormone effects on sex differences in PTSD: The role of heightened sex hormones during trauma. Psychoneuroendocrinology 2019; 109:104416. [PMID: 31472433 DOI: 10.1016/j.psyneuen.2019.104416] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/05/2019] [Accepted: 08/22/2019] [Indexed: 02/08/2023]
Abstract
Women are at least twice as susceptible to developing post-traumatic stress disorder (PTSD) compared to men. Although most research seeking to explain this discrepancy has focussed on the role of oestradiol during fear extinction learning, the role of progesterone has been overlooked, despite relatively consistent findings being reported concerning the role of progesterone during consolidation of emotional and intrusive memories. In this review article, we outline literature supporting the role of progesterone on memory formation, with particular emphasis on potential memory-enhancing properties of progesterone when subjects are placed under stress. It is possible that progesterone directly and indirectly exerts memory-enhancing effects at the time of trauma, which is an effect that may not be necessarily captured during non-stressful paradigms. We propose a model whereby progesterone's steroidogenic relationship to cortisol and brain-derived neurotrophic factor in combination with elevated oestradiol may enhance emotional memory consolidation during trauma and therefore present a specific vulnerability to PTSD formation in women, particularly during the mid-luteal phase of the menstrual cycle.
Collapse
|
155
|
Gururajan A, Reif A, Cryan JF, Slattery DA. The future of rodent models in depression research. Nat Rev Neurosci 2019; 20:686-701. [DOI: 10.1038/s41583-019-0221-6] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2019] [Indexed: 12/15/2022]
|
156
|
Quantitative analysis of Gria1, Gria2, Dlg1 and Dlg4 expression levels in hippocampus following forced swim stress in mice. Sci Rep 2019; 9:14060. [PMID: 31575955 PMCID: PMC6773768 DOI: 10.1038/s41598-019-50689-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/18/2019] [Indexed: 01/09/2023] Open
Abstract
AMPA receptors and interacting proteins are importantly involved in mediating stress-dependent plasticity. Previously we reported that GluA1-containing AMPA receptors and their interaction with PDZ-proteins are required for the experience-dependent expression of behavioral despair in the forced swim test. However, it is unclear if the expression of GluA1-containing AMPA receptors is affected by this type of behavior. Here we investigated in wild type mice, whether hippocampal gene or protein levels of GluA1 or associated PDZ proteins is altered following forced swim stress. We show that expression of Dlg4 (the gene coding for PSD-95) was strongly reduced after two days of forced swimming. In contrast, levels of Dlg1, Gria1, and Gria2 (coding for SAP97, GluA1, and GluA2 respectively) were not affected after one or two days of forced swimming. The changes in gene expression largely did not translate to the protein level. These findings indicate a limited acute effect of forced swim stress on the expression of the investigated targets and suggest that the acute involvement of GluA1-containing AMPA receptors tor forced swim behavior is a result of non-genomic mechanisms.
Collapse
|
157
|
Sex differences in cued fear responses and parvalbumin cell density in the hippocampus following repetitive concussive brain injuries in C57BL/6J mice. PLoS One 2019; 14:e0222153. [PMID: 31487322 PMCID: PMC6728068 DOI: 10.1371/journal.pone.0222153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023] Open
Abstract
There is strong evidence to suggest a link between repeated head trauma and cognitive and emotional disorders, and Repetitive concussive brain injuries (rCBI) may also be a risk factor for depression and anxiety disorders. Animal models of brain injury afford the opportunity for controlled study of the effects of injury on functional outcomes. In this study, male and cycling female C57BL/6J mice sustained rCBI (3x) at 24-hr intervals and were tested in a context and cued fear conditioning paradigm, open field (OF), elevated zero maze and tail suspension test. All mice with rCBI showed less freezing behavior than sham control mice during the fear conditioning context test. Injured male, but not female mice also froze less in response to the auditory cue (tone). Injured mice were hyperactive in an OF environment and spent more time in the open quadrants of the elevated zero maze, suggesting decreased anxiety, but there were no differences between injured mice and sham-controls in depressive-like activity on the tail suspension test. Pathologically, injured mice showed increased astrogliosis in the injured cortex and white matter tracts (optic tracts and corpus callosum). There were no changes in the number of parvalbumin-positive interneurons in the cortex or amygdala, but injured male mice had fewer parvalbumin-positive neurons in the hippocampus. Parvalbumin-reactive interneurons of the hippocampus have been previously demonstrated to be involved in hippocampal-cortical interactions required for memory consolidation, and it is possible memory changes in the fear-conditioning paradigm following rCBI are the result of more subtle imbalances in excitation and inhibition both within the amygdala and hippocampus, and between more widespread brain regions that are injured following a diffuse brain injury.
Collapse
|
158
|
Genario R, de Abreu MS, Giacomini ACVV, Demin KA, Kalueff AV. Sex differences in behavior and neuropharmacology of zebrafish. Eur J Neurosci 2019; 52:2586-2603. [PMID: 31090957 DOI: 10.1111/ejn.14438] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/27/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022]
Abstract
Sex is an important variable in biomedical research. The zebrafish (Danio rerio) is increasingly utilized as a powerful new model organism in translational neuroscience and pharmacology. Mounting evidence indicates important sex differences in zebrafish behavioral and neuropharmacological responses. Here, we discuss the role of sex in zebrafish central nervous system (CNS) models, their molecular mechanisms, recent findings and the existing challenges in this field. We also emphasize the growing utility of zebrafish models in translational neuropharmacological research of sex differences, fostering future CNS drug discovery and the search for novel sex-specific therapies. Finally, we highlight the interplay between sex and environment in zebrafish models of sex-environment correlations as an important strategy of CNS disease modeling using this aquatic organism.
Collapse
Affiliation(s)
- Rafael Genario
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil.,The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, Louisiana
| | - Ana C V V Giacomini
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil.,Postgraduate Program in Environmental Sciences, University of Passo Fundo (UPF), Passo Fundo, Brazil
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.,Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China.,Ural Federal University, Ekaterinburg, Russia
| |
Collapse
|
159
|
Early life stress and the propensity to develop addictive behaviors. Int J Dev Neurosci 2019; 78:156-169. [PMID: 31255718 DOI: 10.1016/j.ijdevneu.2019.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 06/03/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022] Open
Abstract
There is a vast literature on effects of early life manipulations in rodents much of which is aimed at investigating the long-term consequences related to emotion and cognition in adulthood. Less is known about how these manipulations affect responses reflective of alcohol (AUD) and substance (SUD) use disorders. The purpose of this paper is to review the literature of studies that employed early life manipulations and assessed behavioral responses to psychoactive substances, specifically alcohol, opiates, and stimulants, in rodents. While the findings with alcohol are more limited and mixed, studies with opiates and stimulants show strong support for the ability of these manipulations to enhance behavioral responsivity to these substances in line with epidemiological data. Some outcomes show sex differences. The mechanisms that influence these enduring changes may reflect epigenetic alterations. Several studies support a role for altered DNA methylation (and other epigenetic mechanisms) as biological responses to early environmental insults. The chemical changes induced by DNA methylation affect transcriptional activity of DNA and thus can have a long-term impact on the individual's phenotype. Such effects are particularly robust when they occur during sensitive periods of brain development (e.g., first postnatal weeks in rodents). We review this emerging literature as it relates to the known neurobiology of AUDs and SUDs and suggest new avenues of research. Such findings will have implications for the treatment and prevention of AUDs and SUDs and could provide insight into factors that support resiliency.
Collapse
|
160
|
Demin KA, Lakstygal AM, Alekseeva PA, Sysoev M, de Abreu MS, Alpyshov ET, Serikuly N, Wang D, Wang M, Tang Z, Yan D, Strekalova TV, Volgin AD, Amstislavskaya TG, Wang J, Song C, Kalueff AV. The role of intraspecies variation in fish neurobehavioral and neuropharmacological phenotypes in aquatic models. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 210:44-55. [PMID: 30822702 DOI: 10.1016/j.aquatox.2019.02.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 06/09/2023]
Abstract
Intraspecies variation is common in both clinical and animal research of various brain disorders. Relatively well-studied in mammals, intraspecies variation in aquatic fish models and its role in their behavioral and pharmacological responses remain poorly understood. Like humans and mammals, fishes show high variance of behavioral and drug-evoked responses, modulated both genetically and environmentally. The zebrafish (Danio rerio) has emerged as a particularly useful model organism tool to access neurobehavioral and drug-evoked responses. Here, we discuss recent findings and the role of the intraspecies variance in neurobehavioral, pharmacological and toxicological studies utilizing zebrafish and other fish models. We also critically evaluate common sources of intraspecies variation and outline potential strategies to improve data reproducibility and translatability.
Collapse
Affiliation(s)
- Konstantin A Demin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Anton M Lakstygal
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Granov Russian Research Centre of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Polina A Alekseeva
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Maxim Sysoev
- Granov Russian Research Centre of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Murilo S de Abreu
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA; Bioscience Institute, University of Passo Fundo, Passo Fundo, RS, Brazil
| | | | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | - DongMei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - MengYao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - ZhiChong Tang
- School of Pharmacy, Southwest University, Chongqing, China
| | - DongNi Yan
- School of Pharmacy, Southwest University, Chongqing, China
| | - Tatyana V Strekalova
- Department of Neuroscience, Maastricht University, Maastricht, Netherlands; Laboratory of Psychiatric Neurobiology and Department of Normal Physiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | | | - JiaJia Wang
- Research Institute of Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Cai Song
- Research Institute of Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA; Ural Federal University, Ekaterinburg, Russia; ZENEREI Research Center, Slidell, LA, USA; Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Granov Russian Research Centre of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.
| |
Collapse
|
161
|
Dioli C, Patrício P, Sousa N, Kokras N, Dalla C, Guerreiro S, Santos-Silva MA, Rego AC, Pinto L, Ferreiro E, Sotiropoulos I. Chronic stress triggers divergent dendritic alterations in immature neurons of the adult hippocampus, depending on their ultimate terminal fields. Transl Psychiatry 2019; 9:143. [PMID: 31028242 PMCID: PMC6486609 DOI: 10.1038/s41398-019-0477-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/02/2019] [Accepted: 03/08/2019] [Indexed: 12/18/2022] Open
Abstract
Chronic stress, a suggested precipitant of brain pathologies, such as depression and Alzheimer's disease, is known to impact on brain plasticity by causing neuronal remodeling as well as neurogenesis suppression in the adult hippocampus. Although many studies show that stressful conditions reduce the number of newborn neurons in the adult dentate gyrus (DG), little is known about whether and how stress impacts on dendritic development and structural maturation of these newborn neurons. We, herein, demonstrate that chronic stress impacts differentially on doublecortin (DCX)-positive immature neurons in distinct phases of maturation. Specifically, the density of the DCX-positive immature neurons whose dendritic tree reaches the inner molecular layer (IML) of DG is reduced in stressed animals, whereas their dendritic complexity is increased. On the contrary, no change on the density of DCX-positive neurons whose dendritic tree extends to the medial/outer molecular layer (M/OML) of the DG is found under stress conditions, whereas the dendritic complexity of these cells is diminished. In addition, DCX+ cells displayed a more complex and longer arbor in the dendritic compartments located in the granular cell layer of the DG under stress conditions; on the contrary, their dendritic segments localized into the M/OML were shorter and less complex. These findings suggest that the neuroplastic effects of chronic stress on dendritic maturation and complexity of DCX+ immature neurons vary based on the different maturation stage of DCX-positive cells and the different DG sublayer, highlighting the complex and dynamic stress-driven neuroplasticity of immature neurons in the adult hippocampus.
Collapse
Affiliation(s)
- Chrysoula Dioli
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Patrícia Patrício
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Nuno Sousa
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Nikolaos Kokras
- 0000 0001 2155 0800grid.5216.0First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece ,0000 0001 2155 0800grid.5216.0Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- 0000 0001 2155 0800grid.5216.0Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sara Guerreiro
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Miguel A. Santos-Silva
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Ana Cristina Rego
- 0000 0000 9511 4342grid.8051.cCenter for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute of Biochemistry, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - Luísa Pinto
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Elisabete Ferreiro
- 0000 0000 9511 4342grid.8051.cCenter for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute for Interdisciplinary Research of the University of Coimbra (IIIUC), Coimbra, Portugal
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's - PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.
| |
Collapse
|
162
|
Candemir E, Post A, Dischinger US, Palme R, Slattery DA, O'Leary A, Reif A. Limited effects of early life manipulations on sex-specific gene expression and behavior in adulthood. Behav Brain Res 2019; 369:111927. [PMID: 31034851 DOI: 10.1016/j.bbr.2019.111927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 01/19/2023]
Abstract
Exposure to childhood adversity is associated with increased vulnerability to stress-related disorders in adulthood which has been replicated in rodent stress models, whereas environmental enrichment has been suggested to have beneficial effects. However, the exact neurobiological mechanisms underlying these environment influences on adult brain and behavior are not well understood. Therefore, we investigated the long-term effects of maternal separation (MS) or environmental enrichment (EE) in male and female CD1 mice. We found clear sex-specific effects, but limited influence of environmental manipulations, on adult behavior, fecal corticosterone metabolite (FCM) levels and stress- and plasticity related gene expression in discrete brain regions. In detail, adult females displayed higher locomotor activity and FCM levels compared to males and EE resulted in attenuation in both measures, but only in females. There were no sex- or postnatal manipulation-dependent differences in anxiety-related behaviors in either sex. Gene expression analyses revealed that adult males showed higher Fkbp5 mRNA levels in hippocampus, hypothalamus and raphe nuclei, and higher hippocampal Nos1 levels. Interestingly, MS elevated Nos1 levels in hippocampus but reduced Fkbp5 expression in hypothalamus of males. Finally, we also found higher Maoa expression in the hypothalamus of adult females, however no differences were observed in the expression levels of Bdnf, Crhr1, Nr3c1 and Htr1a. Our findings further contribute to sex-dependent differences in behavior, corticosterone and gene expression and reveal that the effects of postnatal manipulations on these parameters in outbred CD1 mice are limited.
Collapse
Affiliation(s)
- Esin Candemir
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Antonia Post
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Germany
| | - Ulrich Severin Dischinger
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Germany
| | - Rupert Palme
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Neuropsychopharmacology, Institute of Psychology, University of Tartu, Tartu, Estonia
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
163
|
Clark JW, Drummond SPA, Hoyer D, Jacobson LH. Sex differences in mouse models of fear inhibition: Fear extinction, safety learning, and fear-safety discrimination. Br J Pharmacol 2019; 176:4149-4158. [PMID: 30710446 DOI: 10.1111/bph.14600] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/26/2018] [Accepted: 01/04/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Women are overrepresented in post-traumatic stress disorder (PTSD), a mental disorder characterised by ineffective inhibition of fear. The use of male animals dominates preclinical studies, which may contribute to a lack of understanding as to why this disparity exists. Thus, the current study explores sex differences in three mouse models of fear inhibition. EXPERIMENTAL APPROACH All experiments tested male and female C57Bl/6J mice. Experiment 1 employed two fear conditioning protocols, in which tones were paired with footshocks of differing intensity (moderate or intense). Fear recall and extinction were tested subsequently. In Experiment 2, safety learning was investigated. Tones were explicitly unpaired with footshocks during safety conditioning. Recall of safety learning was tested 24 hr later. Experiment 3 assessed a model of fear-safety discrimination. Cued stimuli were paired or never paired with footshocks during fear and safety conditioning, respectively. Discrimination between stimuli was assessed 24 hr later. KEY RESULTS In fear extinction, males, compared to females, responded with greater fear in sessions most proximal to conditioning but subsequently showed a more rapid fear extinction over time. Sex differences were not observed during safety learning. During fear-safety discrimination, both males and females discriminated between stimuli; however, males revealed a greater level of freezing to stimuli. CONCLUSION AND IMPLICATIONS The current study provides evidence that sex differences influence fear but not safety-based behaviour in C57Bl/6J mice. These findings indicate that processing of fear, but not safety, may play a greater role in sex differences observed for PTSD. LINKED ARTICLES This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Jacob W Clark
- Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Notting Hill, VIC, Australia.,Department of Pharmacology and Therapeutics, School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Sean P A Drummond
- Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Notting Hill, VIC, Australia
| | - Daniel Hoyer
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Laura H Jacobson
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
164
|
Age and sex differences in the innate defensive behaviors of C57BL/6 mice exhibited in a fear conditioning paradigm and upon exposure to a predatory odor. Physiol Behav 2019; 204:264-274. [PMID: 30840847 DOI: 10.1016/j.physbeh.2019.02.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/23/2019] [Accepted: 02/23/2019] [Indexed: 12/29/2022]
Abstract
Defensive responses of mice include an array of specific behaviors that involve adaptive components based on the assessment of the threat of confrontation. The freezing response is represented by a motionless moment without any specific posture or behavioral sequence, and it is widely used in the fear conditioning paradigms and other relevant defensive situations. However, freezing measurements include fragmental components of several defensive behaviors that are exhibited during the session, such as behavioral inhibition, crouching, and a moment of risk assessment behavior. From an ethological view, behavioral analyses of C57BL/6 mice of both sexes and three different ages (postnatal days (P) 25, 35, and 65) revealed a rich variety of defensive behaviors during a fear conditioning session and in response to predatory odor exposure as a nonconditioned behavior. P-25 and 35 mice exhibited more behavioral inhibition than P-65 adult mice, and P-65 mice exhibited a crouching posture more often than younger mice. This age difference was more pronounced in males. The stretch-attend posture (SAP) increased with age, except in P-25 males, which exhibited robust SAP in response to a conditioned cue; this response indicates that P-25 males are defensive in a situation-nonmatching manner. Situation-dependent defense strategies were revealed in P-35 and 65 mice: Fear conditioning paradigm induced more robust defensive responses than predatory odor exposure, to which mice primarily exhibited SAP. A sex-based difference was revealed in adult mice. Males tended to show more passive defensive responses, such as crouching and withdrawal, and females exhibited more active responses, such as SAP. These age- and sex-based differences may stem from the ethological demands and illustrate adolescent ontogenetic processes of defense behavior.
Collapse
|
165
|
Modelling posttraumatic stress disorders in animals. Prog Neuropsychopharmacol Biol Psychiatry 2019; 90:117-133. [PMID: 30468906 DOI: 10.1016/j.pnpbp.2018.11.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 01/07/2023]
Abstract
Animal models of posttraumatic stress disorder are useful tools to reveal the neurobiological basis of the vulnerability to traumatic events, and to develop new treatment strategies, as well as predicting treatment response contributing to personalized medicine approach. Different models have different construct, face and predictive validity and they model different symptoms of the disease. The most prevalent models are the single prolonged stress, electric foot-shock and predator odor. Freezing as 're-experiencing' in cluster B and startle as 'arousal' in cluster E according to DSM-5 are the most frequently studied parameters; however, several other symptoms related to mood, cognitive and social skills are part of the examinations. Beside behavioral characteristics, symptoms of exaggerated sympathetic activity and hypothalamic-pituitary-adrenocortical axis as well as signs of sleep disturbances are also warranted. Test battery rather than a single test is required to describe a model properly and the results should be interpreted in a comprehensive way, e.g. creating a z-score. Research is shifting to study larger populations and identifying the features of the resilient and vulnerable individuals, which cannot be easily done in humans. Incorporation of the "three hit theory" in animal models may lead to a better animal model of vulnerability and resilience. As women are twice as vulnerable as men, more emphasize should be taken to include female animals. Moreover, hypothesis free testing and big data analysis may help to identify an array of biomarkers instead of a single variable for identification of vulnerability and for the purpose of personalized medicine.
Collapse
|
166
|
Gubert C, Andrejew R, Jacintho Moritz CE, Dietrich F, Vasconcelos-Moreno MP, dos Santos BTMQ, Fijtman A, Kauer-Sant’Anna M, Kapczinski F, da Silva Magalhães PV, Battastini AMO. Bipolar disorder and 1513A>C P2RX7 polymorphism frequency. Neurosci Lett 2019; 694:143-147. [DOI: 10.1016/j.neulet.2018.11.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 10/27/2022]
|
167
|
Wu X, Lv YG, Du YF, Hu M, Reed MN, Long Y, Suppiramaniam V, Hong H, Tang SS. Inhibitory effect of INT-777 on lipopolysaccharide-induced cognitive impairment, neuroinflammation, apoptosis, and synaptic dysfunction in mice. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:360-374. [PMID: 30144494 DOI: 10.1016/j.pnpbp.2018.08.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/02/2018] [Accepted: 08/19/2018] [Indexed: 01/09/2023]
Abstract
Neuroinflammation plays an important role in the pathophysiology of Alzheimer's disease (AD) and memory impairment. Herein, we evaluated the neuroprotective effects of 6-ethyl-23(S)-methyl-cholic acid (INT-777), a specific G-protein coupled bile acid receptor 1 (TGR5) agonist, in the LPS-treated mouse model of acute neurotoxicity. Single intracerebroventricular (i.c.v.) injection of LPS remarkably induced mouse behavioral impairments in Morris water maze, novel object recognition, and Y-maze avoidance tests, which were ameliorated by INT-777 (1.5 or 3.0 μg/mouse, i.c.v.) treatment. Importantly, INT-777 treatment reversed LPS-induced TGR5 down-regulation, suppressed the increase of nuclear NF-κB p65, and mitigated neuroinflammation, evidenced by lower proinflammatory cytokines, less activation of microglia, and increased the ratio of p-CREB/CREB or mBDNF/proBDNF in the hippocampus and frontal cortex. In addition, INT-777 treatment also suppressed neuronal apoptosis, as indicated by the reduction of TUNEL-positive cells, decreased activation of caspase-3, increased the ratio of Bcl-2/Bax, and ameliorated synaptic dysfunction as evidenced by the upregulation of PSD95 and synaptophysin in the hippocampus and frontal cortex. Taken together, this study showed the potential neuroprotective effects of INT-777 against LPS-induced cognitive impairment, neuroinflammation, apoptosis, and synaptic dysfunction in mice.
Collapse
Affiliation(s)
- Xian Wu
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Yang-Ge Lv
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Yi-Feng Du
- Department of Drug Discovery and Development, School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Mei Hu
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Miranda N Reed
- Department of Drug Discovery and Development, School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Yan Long
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China.
| | - Su-Su Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
168
|
Balta G, Dalla C, Kokras N. Women's Psychiatry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1192:225-249. [PMID: 31705497 DOI: 10.1007/978-981-32-9721-0_11] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Brain disorders and mental diseases, in particular, are common and considered as a top global health challenge for the twenty-first century. Interestingly, women suffer more frequently from mental disorders than men. Moreover, women may respond to psychotropic drugs differently than men, and, through their lifespan, they endure sex-orientated social stressors. In this chapter, we present how women may differ in the development and manifestation of mental health issues and how they differ from men in pharmacokinetics and pharmacodynamics. We discuss issues in clinical trials regarding women participation, issues in the use of psychotropic medications in pregnancy, and challenges that psychiatry faces as a result of the wider use of contraceptives, of childbearing at older age, and of menopause. Such issues, among others, demand further women-oriented psychiatric research that can improve the care for women during the course of their lives. Indeed, despite all these known sex differences, psychiatry for both men and women patients uses the same approach. Thereby, a modified paradigm for women's psychiatry, which takes into account all these differences, emerges as a necessity, and psychiatric research should take more vigorously into account sex differences.
Collapse
Affiliation(s)
- Georgia Balta
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece. .,First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
169
|
Aten S, Page CE, Kalidindi A, Wheaton K, Niraula A, Godbout JP, Hoyt KR, Obrietan K. miR-132/212 is induced by stress and its dysregulation triggers anxiety-related behavior. Neuropharmacology 2019; 144:256-270. [PMID: 30342060 PMCID: PMC6823933 DOI: 10.1016/j.neuropharm.2018.10.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/25/2018] [Accepted: 10/14/2018] [Indexed: 02/08/2023]
Abstract
miR-132 and miR-212 are structurally-related microRNAs that are expressed from the same non-coding transcript. Accumulating evidence has shown that the dysregulation of these microRNAs contributes to aberrant neuronal plasticity and gene expression in the mammalian brain. Consistent with this, altered expression of miR-132 is associated with a number of affect-related psychiatric disorders. Here, we tested the functional contribution of the miR-132/212 locus to the development of stress-related and anxiety-like behaviors. Initially, we tested whether expression from the miR-132/212 locus is altered by stress-inducing paradigms. Using a 5-h acute-stress model, we show that both miR-132 and miR-212 are increased more than two-fold in the WT murine hippocampus and amygdala, whereas after a 15 day chronic-stress paradigm, expression of both miR-132 and miR-212 are upregulated more than two-fold within the amygdala but not in the hippocampus. Next, we used a tetracycline-inducible miR-132 overexpression mouse model and a miR-132/212 conditional knockout (cKO) mouse model to examine whether dysregulation of miR-132/212 expression alters basal anxiety-like behaviors. Interestingly, in both the miR-132 overexpression and cKO lines, significant increases in anxiety-like behaviors were detected. Importantly, suppression of transgenic miR-132 expression (via doxycycline administration) mitigated the anxiety-related behaviors. Further, expression of Sirt1 and Pten-two miR-132 target genes that have been implicated in the regulation of anxiety-were differentially regulated in the hippocampus and amygdala of miR-132/212 conditional knockout and miR-132 transgenic mice. Collectively, these data raise the prospect that miR-132 and miR-212 may play a key role in the modulation of stress responsivity and anxiety.
Collapse
Affiliation(s)
- Sydney Aten
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Chloe E Page
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Anisha Kalidindi
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Kelin Wheaton
- Division of Pharmaceutics and Pharmaceutical Chemistry, Ohio State University, Columbus, OH, USA
| | - Anzela Niraula
- Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Jon P Godbout
- Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, Ohio State University, Columbus, OH, USA
| | - Kari R Hoyt
- Division of Pharmaceutics and Pharmaceutical Chemistry, Ohio State University, Columbus, OH, USA
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
170
|
Kokras N, Sotiropoulos I, Besinis D, Tzouveka EL, Almeida OFX, Sousa N, Dalla C. Neuroplasticity-related correlates of environmental enrichment combined with physical activity differ between the sexes. Eur Neuropsychopharmacol 2019; 29:1-15. [PMID: 30497839 DOI: 10.1016/j.euroneuro.2018.11.1107] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 11/01/2018] [Accepted: 11/09/2018] [Indexed: 01/08/2023]
Abstract
Environmental enrichment (EE), comprising positive physical (exercise) and cognitive stimuli, influences neuronal structure and usually improves brain function. The promise of EE as a preventative strategy against neuropsychiatric disease is especially high during early postnatal development when the brain is still amenable to reorganization. Despite the fact that male and female brains differ in terms of connectivity and function that may reflect early life experiences, knowledge of the neural substrates and mechanisms by which such changes arise remains limited. This study compared the impact of EE combined with physical activity on neuroplasticity and its functional consequences in adult male and female rats; EE was provided during the first 3 months of life and our analysis focused on the hippocampus, an area implicated in cognitive behavior as well as the neuroendocrine response to stress. Both male and female rats reared in EE displayed better object recognition memory than their control counterparts. Interestingly, sex differences were revealed in the effects of EE on time spent exploring the objects during this test. Independently of sex, EE increased hippocampal turnover rates of dopamine and serotonin and reduced expression of 5-HT1A receptors; in addition, EE upregulated expression of synaptophysin, a presynaptic protein, in the hippocampus. As compared to their respective controls, EE-exposed males exhibited parallel increases in phosphorylated Tau and the GluN2B receptor, whereas females responded to EE with reduced hippocampal levels of glutamate and GluN2B. Together, these observations provide further evidence on the differential effects of EE on markers of hippocampal neuroplasticity in males and females.
Collapse
Affiliation(s)
- N Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece; First Department of Psychiatry, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - I Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal; Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | - D Besinis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | - E L Tzouveka
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | | | - N Sousa
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - C Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece.
| |
Collapse
|
171
|
Uzuneser TC, Speidel J, Kogias G, Wang AL, de Souza Silva MA, Huston JP, Zoicas I, von Hörsten S, Kornhuber J, Korth C, Müller CP. Disrupted-in-Schizophrenia 1 (DISC1) Overexpression and Juvenile Immune Activation Cause Sex-Specific Schizophrenia-Related Psychopathology in Rats. Front Psychiatry 2019; 10:222. [PMID: 31057438 PMCID: PMC6465888 DOI: 10.3389/fpsyt.2019.00222] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/26/2019] [Indexed: 01/12/2023] Open
Abstract
Synaptic pruning is a critical refinement step during neurodevelopment, and schizophrenia has been associated with overpruning of cortical dendritic spines. Both human studies and animal models implicate disrupted-in-schizophrenia 1 (DISC1) gene as a strong susceptibility factor for schizophrenia. Accumulating evidence supports the involvement of DISC1 protein in the modulation of synaptic elimination during critical periods of neurodevelopment and of dopamine D2-receptor-mediated signaling during adulthood. In many species, synaptic pruning occurs during juvenile and adolescent periods and is mediated by microglia, which can be over-activated by an immune challenge, giving rise to overpruning. Therefore, we sought to investigate possible interactions between a transgenic DISC1 model (tgDISC1) and juvenile immune activation (JIA) by the bacterial cell wall endotoxin lipopolysaccharide on the induction of schizophrenia-related behavioral and neurochemical disruptions in adult female and male rats. We examined possible behavioral aberrations along three major symptom dimensions of schizophrenia including psychosis, social and emotional disruptions, and cognitive impairments. We detected significant gene-environment interactions in the amphetamine-induced locomotion in female animals and in the amphetamine-induced anxiety in male animals. Surprisingly, gene-environment interactions improved social memory in both male and female animals. JIA alone disrupted spatial memory and recognition memory, but only in male animals. DISC1 overexpression alone induced an improvement in sensorimotor gating, but only in female animals. Our neurochemical analyses detected sex- and manipulation-dependent changes in the postmortem monoamine content of animals. Taken together, we here report sex-specific effects of environment and genotype as well as their interaction on behavioral phenotypes and neurochemical profiles relevant for schizophrenia.
Collapse
Affiliation(s)
- Taygun C Uzuneser
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Jil Speidel
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Georgios Kogias
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - An-Li Wang
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, University of Düsseldorf, Düsseldorf, Germany
| | - Maria A de Souza Silva
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, University of Düsseldorf, Düsseldorf, Germany
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, University of Düsseldorf, Düsseldorf, Germany
| | - Iulia Zoicas
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Stephan von Hörsten
- Department of Experimental Therapy, Preclinical Experimental Center, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Carsten Korth
- Department of Neuropathology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
172
|
Let's call the whole thing off: evaluating gender and sex differences in executive function. Neuropsychopharmacology 2019; 44:86-96. [PMID: 30143781 PMCID: PMC6235899 DOI: 10.1038/s41386-018-0179-5] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
The executive functions allow for purposeful, deliberate, and intentional interactions with the world-attention and focus, impulse control, decision making, and working memory. These measures have been correlated with academic outcomes and quality of life, and are impacted by deleterious environmental events throughout the life span, including gestational and early life insults. This review will address the topic of sex differences in executive function including a discussion of differences arising in response to developmental programming. Work on gender differences in human studies and sex differences in animal research will be reviewed. Overall, we find little support for significant gender or sex differences in executive function. An important variable that factors into the interpretation of potential sex differences include differing developmental trajectories. We conclude by discussing future directions for the field and a brief discussion of biological mechanisms.
Collapse
|
173
|
Riparin II ameliorates corticosterone-induced depressive-like behavior in mice: Role of antioxidant and neurotrophic mechanisms. Neurochem Int 2018; 120:33-42. [DOI: 10.1016/j.neuint.2018.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 07/15/2018] [Accepted: 07/21/2018] [Indexed: 02/06/2023]
|
174
|
Gogos A, McCarthy M, Walker AJ, Udawela M, Gibbons A, Dean B, Kusljic S. Differential effects of chronic 17β-oestradiol treatment on rat behaviours relevant to depression. J Neuroendocrinol 2018; 30:e12652. [PMID: 30311279 DOI: 10.1111/jne.12652] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/08/2018] [Accepted: 10/08/2018] [Indexed: 12/18/2022]
Abstract
Sex differences are a prominent feature of the pathophysiology of psychiatric disorders, such as major depressive disorder, which affects women at a higher incidence than men. Research suggests that the most potent endogenous oestrogen, 17β-oestradiol, may have therapeutic potential in treating depression. However, preclinical studies have produced mixed results, likely as a result of various methodological factors such as treatment duration. The present study aimed to investigate the effects of ovariectomy and chronic 17β-oestradiol treatment via a s.c. silastic implant on behaviours relevant to depression in adult female Sprague-Dawley rats. Rats were assessed in the forced swim test, saccharin preference test and novel object recognition memory test, as well as for possible confounding behaviours, including locomotion and anxiety (open field test) and motivation and anxiety (novelty suppressed feeding test). Treatment effects were verified using body and uterus weight, as well as serum concentrations of 17β-oestradiol, progesterone and testosterone. Compared to ovariectomised rats, chronic 17β-oestradiol treatment enhanced saccharin preference and novel object recognition performance. There were no group differences in passive or active coping behaviour when assayed using the forced swim test. Taken together, these results support an antidepressant-like action of oestrogens but highlight that the beneficial effects of chronic 17β-oestradiol treatment may be related to specific depression-related symptoms, particularly anhedonia and memory.
Collapse
Affiliation(s)
- Andrea Gogos
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Madeleine McCarthy
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Adam J Walker
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Madhara Udawela
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Andrew Gibbons
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Brian Dean
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Centre for Mental Health, Faculty of Health, Arts and Design, Swinburne University, Hawthorn, VIC, Australia
| | - Snezana Kusljic
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Nursing, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
175
|
Marchette RCN, Bicca MA, Santos ECDS, de Lima TCM. Distinctive stress sensitivity and anxiety-like behavior in female mice: Strain differences matter. Neurobiol Stress 2018; 9:55-63. [PMID: 30450373 PMCID: PMC6234269 DOI: 10.1016/j.ynstr.2018.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 12/21/2022] Open
Abstract
Epidemiologic studies have shown that the prevalence of stress-related mood disorders is higher in women, which suggests a different response of neuroendocrine circuits involved in the response to stressful events, as well as a genetic background influence. The aim of this study was to investigate the baseline differences in anxiety-like behaviors of females of two commonly used mice strains. Secondly, we have also aimed to study their behavioral and biochemical alterations following stress. Naïve 3-4 months-old Swiss and C57BL/6 female mice were evaluated in the elevated plus maze (EPM) and in the acoustic startle response (ASR) for anxiety-like behaviors. Besides, an independent group of animals from each strain was exposed to cold-restraint stress (30 min/4 °C, daily) for 21 consecutive days and then evaluated in EPM and in the sucrose consumption tests. Twenty-four hours following behavioral experimentation mice were decapitated and their hippocampi (HP) and cortex (CT) dissected for further Western blotting analysis of glucocorticoid receptor (GR) and glial fibrillary acid protein (GFAP). Subsequent to each behavioral protocol, animal blood samples were collected for further plasma corticosterone analysis. C57BL/6 presented a lower anxiety profile than Swiss female mice in both behavioral tests, EPM and ASR. These phenomena could be correlated with the fact that both strains have distinct corticosterone levels and GR expression in the HP at the baseline level. Moreover, C57BL/6 female mice were more vulnerable to the stress protocol, which was able to induce an anhedonic state characterized by lower preference for a sucrose solution. Behavioral anhedonic-like alterations in these animals coincide with reduced plasma corticosterone accompanied with increased GR and GFAP levels, both in the HP. Our data suggest that in C57BL/6 female mice a dysregulation of the hypothalamus-pituitary-adrenal axis (HPA-axis) occurs, in which corticosterone acting on GRs would possibly exert its pro-inflammatory role, ultimately leading to astrocyte activation in response to stress.
Collapse
Affiliation(s)
| | | | | | - Thereza Christina Monteiro de Lima
- Department of Pharmacology, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88049-970, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
176
|
Modulation of the endocannabinoid system by sex hormones: Implications for posttraumatic stress disorder. Neurosci Biobehav Rev 2018; 94:302-320. [DOI: 10.1016/j.neubiorev.2018.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
|
177
|
Goñi-Balentziaga O, Perez-Tejada J, Renteria-Dominguez A, Lebeña A, Labaka A. Social instability in female rodents as a model of stress related disorders: A systematic review. Physiol Behav 2018; 196:190-199. [DOI: 10.1016/j.physbeh.2018.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/29/2018] [Accepted: 09/05/2018] [Indexed: 01/29/2023]
|
178
|
Gender difference in unconditioned and conditioned predator fear responses in Smith's zokors (Eospalax smithii). Glob Ecol Conserv 2018. [DOI: 10.1016/j.gecco.2018.e00503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
179
|
Melancia F, Schiavi S, Servadio M, Cartocci V, Campolongo P, Palmery M, Pallottini V, Trezza V. Sex-specific autistic endophenotypes induced by prenatal exposure to valproic acid involve anandamide signalling. Br J Pharmacol 2018; 175:3699-3712. [PMID: 29968249 DOI: 10.1111/bph.14435] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/05/2018] [Accepted: 06/25/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Autism spectrum disorder (ASD) is more commonly diagnosed in males than in females. Prenatal exposure to the antiepileptic drug valproic acid (VPA) is an environmental risk factor of ASD. Male rats prenatally exposed to VPA show socio-emotional autistic-like dysfunctions that have been related to changes in the activity of the endocannabinoid anandamide. Here, we have investigated if prenatal VPA induced sex-specific autistic endophenotypes involving anandamide signalling. EXPERIMENTAL APPROACH We studied sex-specific differences in the ASD-like socio-emotional, cognitive and repetitive symptoms displayed during development of Wistar rats of both sexes, prenatally exposed to VPA. The involvement of anandamide was followed by Western blotting of cannabinoid CB1 receptors and by inhibiting its metabolism. KEY RESULTS Female rats were less vulnerable to the deleterious effects of prenatal VPA exposure on social communication, emotional reactivity and cognitive performance than male rats. Conversely, as observed in male rats, prenatal VPA exposure induced selective deficits in social play behaviour and stereotypies in the female rat offspring. At the neurochemical level, prenatal VPA exposure altered phosphorylation of CB1 receptors in a sex-specific, age-specific and tissue-specific manner. Enhancing anandamide signalling through inhibition of its degradation reversed the behavioural deficits displayed by VPA-exposed animals of both sexes. CONCLUSIONS AND IMPLICATIONS These findings highlight sexually dimorphic consequences of prenatal VPA exposure that may be related to sex-specific effects of VPA on endocannabinoid neurotransmission in the course of development and introduce a new therapeutic target for reversing autistic-like symptoms in both sexes.
Collapse
Affiliation(s)
- Francesca Melancia
- Department of Science, Section of Biomedical Sciences and Technologies, University 'Roma Tre', Rome, Italy
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, University 'Roma Tre', Rome, Italy
| | - Michela Servadio
- Department of Science, Section of Biomedical Sciences and Technologies, University 'Roma Tre', Rome, Italy
| | - Veronica Cartocci
- Department of Science, Section of Biomedical Sciences and Technologies, University 'Roma Tre', Rome, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology 'V. Erspamer', Sapienza University of Rome, Rome, Italy
| | - Maura Palmery
- Department of Physiology and Pharmacology 'V. Erspamer', Sapienza University of Rome, Rome, Italy
| | - Valentina Pallottini
- Department of Science, Section of Biomedical Sciences and Technologies, University 'Roma Tre', Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University 'Roma Tre', Rome, Italy
| |
Collapse
|
180
|
Najjar F, Ahmad M, Lagace D, Leenen FHH. Sex differences in depression-like behavior and neuroinflammation in rats post-MI: role of estrogens. Am J Physiol Heart Circ Physiol 2018; 315:H1159-H1173. [PMID: 30052050 DOI: 10.1152/ajpheart.00615.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Patients with heart failure (HF) have a high prevalence of depression associated with a worse prognosis, particularly in older women. The present study evaluated whether sex and estrogens affect depression-like behavior and associated neuroinflammation induced by myocardial infarction (MI) in rats. MI was induced by occlusion of the left anterior descending artery in young adult male and female Wistar rats or in ovariectomized (OVX) female rats without and with estrogen [17β-estradiol (E2)] replacement. MI groups showed a comparable degree of cardiac dysfunction. Eight weeks post-MI, male rats with HF exhibited depression-like behaviors, including anhedonia and higher immobility in the sucrose preference and forced swim tests, which were not observed in female rats with HF. In the cued fear conditioning test, male but not female rats with HF froze more than sham rats. After OVX, female sham rats developed mild depression-like behaviors that were pronounced in OVX female rats post-MI and were largely prevented by E2 replacement. Cytokine levels in the plasma and paraventricular nucleus increased in both sexes with HF, but only male rats with HF showed an increase in cytokine levels in the prefrontal cortex. OVX alone did not affect cytokine levels, but OVX-MI caused significant increases in the prefrontal cortex, which were shifted to an anti-inflammatory pattern by E2 replacement. These results suggest that estrogens prevent depression-like behavior induced by HF post-MI in young adult female rats by inhibiting proinflammatory cytokine production and actions in the prefrontal cortex. NEW & NOTEWORTHY In contrast to male rats, female rats with heart failure after myocardial infarction do not develop depression-like behavior or increases in prefrontal cortex cytokines. However, after ovariectomy, female rats exhibit similar changes, which are prevented by 17β-estradiol replacement. Neuroinflammation in the prefrontal cortex in male subjects may contribute to depression-like behavior, whereas its estrogen-dependent absence in female subjects may protect against depression.
Collapse
Affiliation(s)
- Fatimah Najjar
- Brain and Heart Research Group, University of Ottawa Heart Institute , Ottawa, Ontario , Canada
| | - Monir Ahmad
- Brain and Heart Research Group, University of Ottawa Heart Institute , Ottawa, Ontario , Canada
| | - Diane Lagace
- Department of Cellular and Molecular Medicine and Neuroscience Program, University of Ottawa Brain and Mind Institute , Ottawa, Ontario , Canada
| | - Frans H H Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute , Ottawa, Ontario , Canada
| |
Collapse
|
181
|
Gupta P, Uner OE, Nayak S, Grant GR, Kalb RG. SAP97 regulates behavior and expression of schizophrenia risk enriched gene sets in mouse hippocampus. PLoS One 2018; 13:e0200477. [PMID: 29995933 PMCID: PMC6040763 DOI: 10.1371/journal.pone.0200477] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/27/2018] [Indexed: 01/10/2023] Open
Abstract
Synapse associated protein of 97KDa (SAP97) belongs to a family of scaffolding proteins, the membrane-associated guanylate kinases (MAGUKs), that are highly enriched in the postsynaptic density of synapses and play an important role in organizing protein complexes necessary for synaptic development and plasticity. The Dlg-MAGUK family of proteins are structurally very similar, and an effort has been made to parse apart the unique function of each Dlg-MAGUK protein by characterization of knockout mice. Knockout mice have been generated and characterized for PSD-95, PSD-93, and SAP102, however SAP97 knockout mice have been impossible to study because the SAP97 null mice die soon after birth due to a craniofacial defect. We studied the transcriptomic and behavioral consequences of a brain-specific conditional knockout of SAP97 (SAP97-cKO). RNA sequencing from hippocampi from control and SAP97-cKO male animals identified 67 SAP97 regulated transcripts. As large-scale genetic studies have implicated MAGUKs in neuropsychiatric disorders such as intellectual disability, autism spectrum disorders, and schizophrenia (SCZ), we analyzed our differentially expressed gene (DEG) set for enrichment of disease risk-associated genes, and found our DEG set to be specifically enriched for SCZ-related genes. Subjecting SAP97-cKO mice to a battery of behavioral tests revealed a subtle male-specific cognitive deficit and female-specific motor deficit, while other behaviors were largely unaffected. These data suggest that loss of SAP97 may have a modest contribution to organismal behavior. The SAP97-cKO mouse serves as a stepping stone for understanding the unique role of SAP97 in biology.
Collapse
Affiliation(s)
- Preetika Gupta
- Neuroscience Graduate Group, Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ogul E. Uner
- School of Arts and Sciences, Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Soumyashant Nayak
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gregory R. Grant
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Robert G. Kalb
- Feinberg School of Medicine, Department of Neurology, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
182
|
Harris EP, Allardice HA, Schenk AK, Rissman EF. Effects of maternal or paternal bisphenol A exposure on offspring behavior. Horm Behav 2018; 101:68-76. [PMID: 28964733 PMCID: PMC5882611 DOI: 10.1016/j.yhbeh.2017.09.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/14/2017] [Accepted: 09/25/2017] [Indexed: 12/15/2022]
Abstract
Bisphenol A (BPA) is an endocrine disrupting chemical used in the production of polycarbonate plastics and resins. Exposure to BPA during gestation has been proposed as a risk factor for the development of neurobehavioral disorders, such as autism spectrum disorder. To address the behavioral impact of developmental exposure to BPA, we tested offspring of mice exposed to a daily low dose of BPA during pregnancy. We also asked if preconception exposure of the sire affected behaviors in offspring. Sires that consumed BPA for 50days prior to mating weighed less than controls, but no effects on any reproductive measures were noted. Juvenile offspring exposed to BPA maternally, but not paternally, spent less time in the open arms of the elevated plus maze than controls, indicating increased anxiety-like behavior. However, neither parental exposure group differed significantly from controls in the social recognition task. We also assessed the behaviors of maternally exposed offspring in two novel tasks: ultrasonic vocalizations (USVs) in pups and operant reversal learning in adults. Maternal BPA exposure increased the duration and median frequency of USVs emitted by pups during maternal separation. In the reversal learning task, females responded more accurately and earned more rewards than males. Additionally, control females received more rewards than BPA females during the acquisition phase of the task. These are among the first studies conducted to ask if BPA exposure via the sire affects offspring behavior and the first study to report effects of gestational BPA exposure on pup USVs and adult operant responding.
Collapse
Affiliation(s)
- Erin P Harris
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| | - Heather A Allardice
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| | - A Katrin Schenk
- Department of Physics, Randolph College, Lynchburg, VA 24503, USA
| | - Emilie F Rissman
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
183
|
Zucker I. Psychoactive drug exposure during breastfeeding: a critical need for preclinical behavioral testing. Psychopharmacology (Berl) 2018; 235:1335-1346. [PMID: 29549392 DOI: 10.1007/s00213-018-4873-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/26/2018] [Indexed: 12/11/2022]
Abstract
Breastfeeding women are excluded from clinical trials of psychoactive drugs because of ethical concerns. Animal testing, which often is predictive of adverse effects in humans, represents the only avenue available for assessing drug safety for human offspring exposed to drugs during lactation. I determined whether behavioral outcomes for children exposed during breastfeeding to antidepressants, anxiolytics, antipsychotics, anti-seizure medications, analgesics, sedatives, and marijuana can be predicted by rodent studies of offspring exposed to drugs during lactation. Animal data were available for only 10 of 80 CNS-active drugs canvassed. Behavioral deficits in adolescence or adulthood in rats and mice after various drug exposures during lactation included reductions in sexual behavior, increased anxiety, hyperactivity, and impaired learning and memory. Whether similar adverse effects will emerge in adulthood in children exposed to drugs during breastfeeding is unknown. Rodent research has the potential to forecast impairments in breastfed children long before information emerges from post-marketing reports and should be prioritized during preclinical drug evaluation by the FDA for new drugs and for drugs currently prescribed off-label for lactating women.
Collapse
Affiliation(s)
- Irving Zucker
- Departments of Psychology and Integrative Biology, University of California, Berkeley, CA, 94720, USA. .,Psychology Department, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
184
|
Abstract
OBJECTIVE Animal models are frequently used to examine stress response, but experiments seldom include females. The connection between the microbiota-gut-brain axis and behavioral stress response is investigated here using a mixed-sex mouse cohort. METHODS CF-1 mice underwent alternating days of restraint and forced swim for 19 days (male n = 8, female n = 8) with matching numbers of control animals at which point the 16S rRNA genes of gut microbiota were sequenced. Mixed linear models accounting for stress status and sex with individuals nested in cage to control for cage effects evaluated these data. Murine behaviors in elevated plus-maze, open-field, and light/dark box were investigated. RESULTS Community-level associations with sex, stress, and their interaction were significant. Males had higher microbial diversity than females (p = .025). Of the 638 operational taxonomic units detected in at least 25% of samples, 94 operational taxonomic units were significant: 31 (stress), 61 (sex), and 34 (sex-stress interaction). Twenty of the 39 behavioral measures were significant for stress, 3 for sex, and 6 for sex-stress. However, no significant associations between behavioral measures and specific microbes were detected. CONCLUSIONS These data suggest sex influences stress response and the microbiota-gut-brain axis and that studies of behavior and the microbiome therefore benefit from consideration of how sex differences drive behavior and microbial community structure. Host stress resilience and absence of associations between stress-induced behaviors with specific microbes suggests that hypothalamic-pituitary-adrenal axis activation represents a threshold for microbial influence on host behavior. Future studies are needed in examining the intersection of sex, stress response, and the microbiota-gut-brain axis.
Collapse
|
185
|
Harris AZ, Atsak P, Bretton ZH, Holt ES, Alam R, Morton MP, Abbas AI, Leonardo ED, Bolkan SS, Hen R, Gordon JA. A Novel Method for Chronic Social Defeat Stress in Female Mice. Neuropsychopharmacology 2018; 43:1276-1283. [PMID: 29090682 PMCID: PMC5916350 DOI: 10.1038/npp.2017.259] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/18/2017] [Accepted: 10/24/2017] [Indexed: 01/14/2023]
Abstract
Historically, preclinical stress studies have often omitted female subjects, despite evidence that women have higher rates of anxiety and depression. In rodents, many stress susceptibility and resilience studies have focused on males as one commonly used paradigm-chronic social defeat stress-has proven challenging to implement in females. We report a new version of the social defeat paradigm that works in female mice. By applying male odorants to females to increase resident male aggressive behavior, we find that female mice undergo repeated social defeat stress and develop social avoidance, decreased sucrose preference, and decreased time in the open arms of the elevated plus maze relative to control mice. Moreover, a subset of the female mice in this paradigm display resilience, maintaining control levels of social exploration and sucrose preference. This method produces comparable results to those obtained in male mice and will greatly facilitate studying female stress susceptibility.
Collapse
Affiliation(s)
- Alexander Z Harris
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
| | - Piray Atsak
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Zachary H Bretton
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Emma S Holt
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Raisa Alam
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Mitchell P Morton
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
| | - Atheir I Abbas
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
| | - E David Leonardo
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
| | - Scott S Bolkan
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - René Hen
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
| | - Joshua A Gordon
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
| |
Collapse
|
186
|
Schreurs BG, Smith-Bell C, Burhans LB. Sex differences in a rabbit eyeblink conditioning model of PTSD. Neurobiol Learn Mem 2018; 155:519-527. [PMID: 29702205 DOI: 10.1016/j.nlm.2018.04.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/12/2018] [Accepted: 04/22/2018] [Indexed: 01/18/2023]
Abstract
We have developed a rabbit model of posttraumatic stress disorder (PTSD) which recapitulates several core features of PTSD, particularly hyperarousal and conditioned responding to trauma-associated cues. The work conducted with this model has all been done in male rabbits and, given sex differences in PTSD prevalence, it is important to expand our animal model of PTSD to include female rabbits to determine if they develop core features of PTSD, and if those core features can be treated. This is particularly important because, contrary to human studies, nearly all animal studies have found that males are consistently more vulnerable to various forms of acute and chronic stress than females. Using eyeblink conditioning in which we paired tone with a brief periorbital shock, we found that although both male and female rabbits acquired identical levels of conditioning, females showed more hyperarousal after conditioning but seemed to respond somewhat better to treatment.
Collapse
Affiliation(s)
- Bernard G Schreurs
- Blanchette Rockefeller Neurosciences Institute, West Virginia University, United States; Department of Physiology, Pharmacology and Neuroscience, West Virginia University, United States.
| | - Carrie Smith-Bell
- Blanchette Rockefeller Neurosciences Institute, West Virginia University, United States; Department of Physiology, Pharmacology and Neuroscience, West Virginia University, United States
| | - Lauren B Burhans
- Blanchette Rockefeller Neurosciences Institute, West Virginia University, United States; Department of Physiology, Pharmacology and Neuroscience, West Virginia University, United States
| |
Collapse
|
187
|
Lorsch ZS, Loh YHE, Purushothaman I, Walker DM, Parise EM, Salery M, Cahill ME, Hodes GE, Pfau ML, Kronman H, Hamilton PJ, Issler O, Labonté B, Symonds AE, Zucker M, Zhang TY, Meaney MJ, Russo SJ, Shen L, Bagot RC, Nestler EJ. Estrogen receptor α drives pro-resilient transcription in mouse models of depression. Nat Commun 2018; 9:1116. [PMID: 29549264 PMCID: PMC5856766 DOI: 10.1038/s41467-018-03567-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 02/22/2018] [Indexed: 11/30/2022] Open
Abstract
Most people exposed to stress do not develop depression. Animal models have shown that stress resilience is an active state that requires broad transcriptional adaptations, but how this homeostatic process is regulated remains poorly understood. In this study, we analyze upstream regulators of genes differentially expressed after chronic social defeat stress. We identify estrogen receptor α (ERα) as the top regulator of pro-resilient transcriptional changes in the nucleus accumbens (NAc), a key brain reward region implicated in depression. In accordance with these findings, nuclear ERα protein levels are altered by stress in male and female mice. Further, overexpression of ERα in the NAc promotes stress resilience in both sexes. Subsequent RNA-sequencing reveals that ERα overexpression in NAc reproduces the transcriptional signature of resilience in male, but not female, mice. These results indicate that NAc ERα is an important regulator of pro-resilient transcriptional changes, but with sex-specific downstream targets.
Collapse
Affiliation(s)
- Zachary S Lorsch
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Yong-Hwee Eddie Loh
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Deena M Walker
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Eric M Parise
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Marine Salery
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Georgia E Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| | - Madeline L Pfau
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Hope Kronman
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Peter J Hamilton
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Orna Issler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Benoit Labonté
- Department of Neuroscience and Psychiatry, Faculty of Medicine, Laval University, 2601 Chemin de la Canardière Québec, QC, G1J 2G3, Canada
| | - Ann E Symonds
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Matthew Zucker
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Tie Yuan Zhang
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Institute, Sackler Program for Epigenetics and Psychobiology, Departments of Psychiatry and Neurology and Neurosurgery, McGill University, 6875 Boulevard Lasalle, Montréal, QC, H4H 1R3, Canada
| | - Michael J Meaney
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Institute, Sackler Program for Epigenetics and Psychobiology, Departments of Psychiatry and Neurology and Neurosurgery, McGill University, 6875 Boulevard Lasalle, Montréal, QC, H4H 1R3, Canada
| | - Scott J Russo
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Li Shen
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA
| | - Rosemary C Bagot
- Departments of Psychology & Psychiatry, Ludmer Centre for Neuroinformatics and Mental Health, McGill University, 1205 Avenue Dr Penfield, Montréal, QC, H3A 1B1, Canada
| | - Eric J Nestler
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, New York, NY, 10029, USA.
| |
Collapse
|
188
|
Singh G, Singh V, Sobolewski M, Cory-Slechta DA, Schneider JS. Sex-Dependent Effects of Developmental Lead Exposure on the Brain. Front Genet 2018; 9:89. [PMID: 29662502 PMCID: PMC5890196 DOI: 10.3389/fgene.2018.00089] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/02/2018] [Indexed: 11/23/2022] Open
Abstract
The role of sex as an effect modifier of developmental lead (Pb) exposure has until recently received little attention. Lead exposure in early life can affect brain development with persisting influences on cognitive and behavioral functioning, as well as, elevated risks for developing a variety of diseases and disorders in later life. Although both sexes are affected by Pb exposure, the incidence, manifestation, and severity of outcomes appears to differ in males and females. Results from epidemiologic and animal studies indicate significant effect modification by sex, however, the results are not consistent across studies. Unfortunately, only a limited number of human epidemiological studies have included both sexes in independent outcome analyses limiting our ability to draw definitive conclusions regarding sex-differentiated outcomes. Additionally, due to various methodological differences across studies, there is still not a good mechanistic understanding of the molecular effects of lead on the brain and the factors that influence differential responses to Pb based on sex. In this review, focused on prenatal and postnatal Pb exposures in humans and animal models, we discuss current literature supporting sex differences in outcomes in response to Pb exposure and explore some of the ideas regarding potential molecular mechanisms that may contribute to sex-related differences in outcomes from developmental Pb exposure. The sex-dependent variability in outcomes from developmental Pb exposure may arise from a combination of complex factors, including, but not limited to, intrinsic sex-specific molecular/genetic mechanisms and external risk factors including sex-specific responses to environmental stressors which may act through shared epigenetic pathways to influence the genome and behavioral output.
Collapse
Affiliation(s)
- Garima Singh
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Vikrant Singh
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Jay S Schneider
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
189
|
Abstract
Several recent studies have claimed that rodents have good visual recognition abilities. However, the extent to which rats can recognize other rats and distinguish between males and females using visual information alone remains unclear. In the present study, we investigated the ability of rats to visually recognize mirror, video-recorded, and still images and to discriminate between images of males and females. Rats were tested in a place preference apparatus with a mirror, a video-recorded image of a rat, or a still image of a rat at one end. The data were assessed using t-test with Bonferroni correction. Male and female rats spent significantly more time in the mirror chamber and the video-recorded image chamber than in their respective blank chambers (P < 0.05), and male rats also spent more time in the chamber containing a still image. Furthermore, it was found that male rats exhibited significantly more sniffing behavior around the mirror than in the blank chamber (P < 0.05), whereas female rats were no significant differences in the sniffing behaviors in the mirror, moving or still image experiments. Identical results were obtained regardless of whether the rat in the image was the same or opposite sex. These results indicate that rats can process the differences in mirror, video-recorded, and still images as visual information, but are unable to use this information to distinguish between the sexes.
Collapse
|
190
|
Ladha S, Qiu X, Casal L, Caron NS, Ehrnhoefer DE, Hayden MR. Constitutive ablation of caspase-6 reduces the inflammatory response and behavioural changes caused by peripheral pro-inflammatory stimuli. Cell Death Discov 2018; 4:40. [PMID: 29560279 PMCID: PMC5849887 DOI: 10.1038/s41420-018-0043-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 02/12/2018] [Indexed: 12/22/2022] Open
Abstract
Traditionally, the family of caspases has been subcategorised according to their respective main roles in mediating apoptosis or inflammation. However, recent studies have revealed that caspases participate in diverse cellular functions beyond their canonical roles. Caspase-6 (C6) is one such protease known for its role as a pro-apoptotic executioner caspase and its aberrant activity in several neurodegenerative diseases. In addition to apoptosis, C6 has been shown to regulate B-cell activation and differentiation in plasma cells as well as macrophage activation. Furthermore, C6 has recently been postulated to play a role in mediating the inflammatory response through the production of TNF-α. In this study we further examine the role of C6 in mediating the inflammatory response and its contribution to the manifestation of behavioural abnormalities in mice. We find that C6 is a positive regulator of TNF-α transcription in macrophages and that ablation of C6 reduces lipopolysaccharide (LPS)-induced TNF-α levels in plasma. Furthermore, loss of C6 attenuates LPS-induced behavioural changes in mice and protects neurons from cytokine-mediated toxicity. These data further support the involvement of C6 in the inflammatory response and point to a previously unknown role for C6 in the pathophysiology of depression.
Collapse
Affiliation(s)
- Safia Ladha
- 1Centre for Molecular Medicine and Therapeutics (CMMT), CFRI, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC V5Z 4H4 Canada
| | - Xiaofan Qiu
- 1Centre for Molecular Medicine and Therapeutics (CMMT), CFRI, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC V5Z 4H4 Canada
| | - Lorenzo Casal
- 1Centre for Molecular Medicine and Therapeutics (CMMT), CFRI, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC V5Z 4H4 Canada
| | - Nicholas S Caron
- 1Centre for Molecular Medicine and Therapeutics (CMMT), CFRI, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC V5Z 4H4 Canada
| | - Dagmar E Ehrnhoefer
- 1Centre for Molecular Medicine and Therapeutics (CMMT), CFRI, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC V5Z 4H4 Canada.,Present Address: BioMed X Innovation Center, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Michael R Hayden
- 1Centre for Molecular Medicine and Therapeutics (CMMT), CFRI, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC V5Z 4H4 Canada
| |
Collapse
|
191
|
The Effect of Gentle Handling on Depressive-Like Behavior in Adult Male Mice: Considerations for Human and Rodent Interactions in the Laboratory. Behav Neurol 2018; 2018:2976014. [PMID: 29692869 PMCID: PMC5859797 DOI: 10.1155/2018/2976014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/08/2017] [Accepted: 12/19/2017] [Indexed: 02/04/2023] Open
Abstract
Environmental factors play a significant role in well-being of laboratory animals. Regulations and guidelines recommend, if not require, that stressors such as bright lighting, smells, and noises are eliminated or reduced to maximize animal well-being. A factor that is often overlooked is handling and how researchers interact with their animals. Researchers, lab assistants, and husbandry staff in animal facilities may use inconsistent handling methods when interacting with rodents, but humans should be considered a part of the animal's social environment. This study examined the effects of different handling techniques on depressive-like behavior, measured by the Porsolt forced swim test, in adult C57BL/6J male mice. The same two researchers handled the mice in a gentle, aggressive, or minimal (control) fashion over approximately two weeks prior to testing. The results demonstrated a beneficial effect of gentle handling: gentle handling reduced swimming immobility in the forced swim test compared to mice that were aggressively or minimally handled. We argue that gentle handling, rather than methodical handling, can foster a better relationship between the handlers and rodents. Although handling is not standardized across labs, consistent gentle handling allows for less challenging behavioral testing, better data collection, and overall improved animal welfare.
Collapse
|
192
|
Wright KN, Kabbaj M. Sex differences in sub-anesthetic ketamine's antidepressant effects and abuse liability. Curr Opin Behav Sci 2018; 23:36-41. [PMID: 30038955 DOI: 10.1016/j.cobeha.2018.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sub-anesthetic ketamine produces rapid antidepressant effects in patients with bipolar and unipolar major depression where conventional monoaminergic-based antidepressant drugs have been ineffective or ridden with side effects. A single ketamine infusion can produce antidepressant effects lasting up to two weeks, and multiple ketamine infusions prolong this effect. Pre-clinical studies are underway to uncover ketamine's mechanisms of action, but there are still many questions unanswered regarding the safety of its long-term use. Abuse liability is one area of concern, as recreational ketamine use is an ongoing issue in many parts of the world. Another understudied area is sex differences in responsivity to ketamine. Women are twice as likely as men to be diagnosed with depression, and they progress through stages of drug addiction more rapidly than their male counterparts. Despite this, preclinical studies in ketamine's antidepressant and addictive-like behaviors in females are limited. These intersecting factors in recent clinical and pre-clinical studies are reviewed to characterize ketamine's therapeutic potential, its limitations, and its potential mechanisms of action.
Collapse
Affiliation(s)
- Katherine N Wright
- Florida State University, Department of Biomedical Sciences, Program in Neuroscience, Tallahassee, FL
| | - Mohamed Kabbaj
- Florida State University, Department of Biomedical Sciences, Program in Neuroscience, Tallahassee, FL
| |
Collapse
|
193
|
Chrousos GP, Pervanidou P, Dalla C. Mini-reviews based on the First Conference of the Institute of Stress Biology & Medicine "Systems Biology-Medicine and Stress". Hormones (Athens) 2018; 17:3-4. [PMID: 29858863 DOI: 10.1007/s42000-018-0027-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- George P Chrousos
- First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota Pervanidou
- First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
- Division of Developmental and Behavioral Pediatrics, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece.
| | - Christina Dalla
- Department of Pharmacology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
194
|
Alimohammadi S, Hosseini MS, Behbood L. Prenatal Exposure to Zinc Oxide Nanoparticles Can Induce Depressive-Like Behaviors in Mice Offspring. Int J Pept Res Ther 2018. [DOI: 10.1007/s10989-018-9686-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
195
|
Anderson E, McWaters M, McFadden L, Matuszewich L. Defensive burying as an ethological approach to studying anxiety: Influence of juvenile methamphetamine on adult defensive burying behavior in rats. LEARNING AND MOTIVATION 2018. [DOI: 10.1016/j.lmot.2017.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
196
|
Dadomo H, Gioiosa L, Cigalotti J, Ceresini G, Parmigiani S, Palanza P. What is stressful for females? Differential effects of unpredictable environmental or social stress in CD1 female mice. Horm Behav 2018; 98:22-32. [PMID: 29187314 DOI: 10.1016/j.yhbeh.2017.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/14/2017] [Accepted: 11/21/2017] [Indexed: 01/01/2023]
Abstract
Stressful life events are a major factor in the etiology of several diseases, such as cardiovascular, inflammatory and psychiatric disorders (i.e., depression and anxiety), with the two sexes greatly differing in vulnerability. In humans and other animals, physiological and behavioral responses to stress are strongly dependent on gender, and conditions that are stressful for males are not necessarily stressful for females. Hence the need of an animal model of social chronic stress specifically designed for females. In the present study we aimed to compare the effects of two different chronic stress procedures in female mice, by investigating the impact of 4weeks of nonsocial unpredictable, physical stress by the Chronic Mild Stress paradigm (CMS; Exp.1) or of Social Instability Stress (SIS; Exp.2) on physiological, endocrine and behavioral parameters in adult female mice. CMS had a pronounced effect on females' response to novelty (i.e., either novel environment or novel social stimulus), body weight growth and hormonal profile. Conversely, 4weeks of social instability did not alter females' response to novelty nor hormonal levels but induced anhedonia. Our findings thus showed that female mice were more sensitive to nonsocial stress due to unpredictable physical environment than to social instability stressors. Neither of these stress paradigms, however, induced a consistent behavioral and physiological stress response in female mice comparable to that induced by chronic stress procedures in male mice, thus confirming the difficulties of developing a robust and validated model of chronic psychosocial stress in female mice.
Collapse
Affiliation(s)
- Harold Dadomo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Laura Gioiosa
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Jenny Cigalotti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Graziano Ceresini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Parmigiani
- Department of Chemistry, Life Sciences and Environmental Sustainaibility, University of Parma, Parma, Italy
| | - Paola Palanza
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| |
Collapse
|
197
|
Bolea-Alamanac B, Bailey SJ, Lovick TA, Scheele D, Valentino R. Female psychopharmacology matters! Towards a sex-specific psychopharmacology. J Psychopharmacol 2018; 32:125-133. [PMID: 29405799 DOI: 10.1177/0269881117747578] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There is increasing recognition that women have a higher prevalence of certain psychiatric illnesses, and a differential treatment response and course of illness compared to men. Additionally, clinicians deal with a number of disorders like premenstrual syndrome, premenstrual dysphoric disorder, and postpartum depression, which affect women specifically and for which treatment and biological pathways are still unclear. In this article we highlight recent research which suggests that different biological mechanisms may underlie sex differences in responsiveness to stress. Sex differences are evident at the receptor level; where the corticotropin-releasing factor receptor shows differential coupling to adaptor proteins in males and females. The neuropeptide oxytocin also shows sex-specific effects in a range of social behaviors. It may act as a biomarker in post-traumatic stress disorder where sex differences are evident. Studies in women using hormonal contraception show that some of these oxytocin-mediated effects are likely influenced by sex hormones. In female rats rapid changes in circulating progesterone levels are associated with exaggerated behavioral responses to mild stress and blunted responses to benzodiazepines that could be prevented by acute treatment with low-dose fluoxetine. Perceived barriers in research on women have hindered progress. The development of a sex-specific psychopharmacology as a basis for translating this type of research into clinical practice is vital to improve treatment outcomes for women.
Collapse
Affiliation(s)
| | - Sarah J Bailey
- 2 Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Thelma A Lovick
- 3 School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Dirk Scheele
- 4 Division of Medical Psychology, University of Bonn Medical Center, Bonn, Germany
| | - Rita Valentino
- 5 Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
198
|
Kokras N, Pastromas N, Papasava D, de Bournonville C, Cornil CA, Dalla C. Sex differences in behavioral and neurochemical effects of gonadectomy and aromatase inhibition in rats. Psychoneuroendocrinology 2018; 87:93-107. [PMID: 29054014 DOI: 10.1016/j.psyneuen.2017.10.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 10/05/2017] [Accepted: 10/09/2017] [Indexed: 01/21/2023]
Abstract
Aromatase inhibitors, which are widely used for the treatment of estrogen-dependent cancers, have been associated with psychiatric side effects ranging from mania to depression. In the present study, we investigated sex differences in the behavioral and neurochemical effects of aromatase inhibition on male and female, sham-operated or gonadectomized adult rats. Three weeks after surgery, rats received chronic treatment with the aromatase inhibitor letrozole or vehicle and were then subjected to the open field test, which assesses general activity. Half of the subjects were subsequently exposed to the stressful procedure of the forced swim test (FST), which is also a test of antidepressant activity. Aromatase activity was analyzed in the hypothalamus and testosterone and corticosterone were assayed in the blood serum of all rats. The hippocampus and prefrontal cortex (PFC) were analyzed for monoamine (noradrenaline, dopamine and serotonin), as well as amino acid (GABA, glutamate, glycine, taurine, alanine and histidine) levels. The observed decrease in hypothalamic aromatase activity confirmed the efficacy of letrozole treatment in both sexes. Moreover, letrozole enhanced testosterone levels in sham-operated females. In the open field test, females were overall more active and explorative than males and gonadectomy eliminated this sex difference. In the FST, females exhibited overall higher immobility than males and gonadectomy further enhanced this passive behavior in both sexes. However, sustained aromatase inhibition had no effect on open field and FST behaviors. Head shakes during FST, which were fewer in females than in males, were reduced by castration in males and by letrozole treatment in ovariectomized females, suggesting a role of testosterone and extra-gonadal estrogens in the expression of this behavior. Sustained aromatase inhibition also decreased noradrenaline and the dopaminergic turnover rates [DOPAC/DA, HVA/DA] in the hippocampus and PFC of male and female rats, irrespectively of gonadectomy. Moreover, letrozole treatment enhanced the serotonergic turnover [5HIAA/5HT] rate in the hippocampus of males and females, irrespectively of gonadectomy. Amino acid levels were not influenced by letrozole, but sex differences were demonstrated with higher levels in the PFC of females vs. males. Present findings suggest that the neuropsychiatric effects of aromatase inhibition can be attributed to the inhibition of extragonadal estrogen synthesis, presumably in the brain, and could be further associated with serotonergic and catecholaminergic changes in brain regions involved in mood and cognition. Importantly, present data could be linked with the neurobiology of affective side-effects in post-menopausal women receiving aromatase inhibitors.
Collapse
Affiliation(s)
- N Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece; First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - N Pastromas
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece
| | - D Papasava
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece
| | - C de Bournonville
- Behavioral Neuroendocrinology Research Group, GIGA Neurosciences, University of Liège, Belgium
| | - C A Cornil
- Behavioral Neuroendocrinology Research Group, GIGA Neurosciences, University of Liège, Belgium
| | - C Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
199
|
Kafetzopoulos V, Kokras N, Sotiropoulos I, Oliveira JF, Leite-Almeida H, Vasalou A, Sardinha VM, Papadopoulou-Daifoti Z, Almeida OFX, Antoniou K, Sousa N, Dalla C. The nucleus reuniens: a key node in the neurocircuitry of stress and depression. Mol Psychiatry 2018; 23:579-586. [PMID: 28397837 PMCID: PMC5822458 DOI: 10.1038/mp.2017.55] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 02/08/2023]
Abstract
The hippocampus and prefrontal cortex (PFC) are connected in a reciprocal manner: whereas the hippocampus projects directly to the PFC, a polysynaptic pathway that passes through the nucleus reuniens (RE) of the thalamus relays inputs from the PFC to the hippocampus. The present study demonstrates that lesioning and/or inactivation of the RE reduces coherence in the PFC-hippocampal pathway, provokes an antidepressant-like behavioral response in the forced swim test and prevents, but does not ameliorate, anhedonia in the chronic mild stress (CMS) model of depression. Additionally, RE lesioning before CMS abrogates the well-known neuromorphological and endocrine correlates of CMS. In summary, this work highlights the importance of the reciprocal connectivity between the hippocampus and PFC in the establishment of stress-induced brain pathology and suggests a role for the RE in promoting resilience to depressive illness.
Collapse
Affiliation(s)
- V Kafetzopoulos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - N Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece,First Department of Psychiatry, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - I Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal,ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
| | - J F Oliveira
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal,ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
| | - H Leite-Almeida
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal,ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
| | - A Vasalou
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - V M Sardinha
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal,ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
| | - Z Papadopoulou-Daifoti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - O F X Almeida
- NeuroAdaptations Group, Max Planck Institute of Psychiatry, Munich, Germany
| | - K Antoniou
- Department of Pharmacology, University of Ioannina, Ioannina, Greece
| | - N Sousa
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal,ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
| | - C Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece,Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece. E-mail:
| |
Collapse
|
200
|
Freudenberg F, O’Leary A, Aguiar DC, Slattery DA. Challenges with modelling anxiety disorders: a possible hindrance for drug discovery. Expert Opin Drug Discov 2017; 13:279-281. [DOI: 10.1080/17460441.2018.1418321] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Aet O’Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
- Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Tartu, Estonia
| | - Daniele C. Aguiar
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - David A. Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|