1
|
Zhong Q, Lai S, He J, Zhong S, Song X, Wang Y, Zhang Y, Chen G, Yan S, Jia Y. Gender-related alterations of serum trace elements and neurometabolism in the anterior cingulate cortex of patients with major depressive disorder. J Affect Disord 2024; 360:176-187. [PMID: 38723680 DOI: 10.1016/j.jad.2024.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND It is widely known that sex differences have a significant impact on patients with major depressive disorder (MDD). This study aims to evaluate the sex-related connection between serum trace elements and changes in neurometabolism in the anterior cingulate cortex (ACC) of MDD patients. METHODS 109 untreated MDD patients and 59 healthy controls underwent proton magnetic resonance spectroscopy (1H-MRS) under resting conditions. We measured metabolic ratios in the ACC from both sides. Additionally, venous blood samples were taken from all participants to detect calcium (Ca), phosphorus, magnesium (Mg), copper (Cu), ceruloplasmin (CER), zinc (Zn), and iron (Fe) levels. We performed association and interaction analyses to explore the connections between the disease and gender. RESULTS In individuals with MDD, the Cu/Zn ratio increased, while the levels of Mg, CER, Zn and Fe decreased. Male MDD patients had lower Cu levels, while female patients had an increased Cu/Zn ratio. We observed significant gender differences in Cu, CER and the Cu/Zn ratio in MDD. Male patients showed a reduced N-acetyl aspartate (NAA)/phosphocreatine + creatine (PCr + Cr) ratio in the left ACC. The NAA/PCr + Cr ratio decreased in the right ACC in patients with MDD. In the left ACC of male MDD patients, the Cu/Zn ratio was inversely related to the NAA/PCr + Cr ratio, and Fe levels were negatively associated with the GPC + PC/PCr + Cr ratio. CONCLUSIONS Our findings highlight gender-specific changes in Cu homeostasis among male MDD patients. The Cu/Zn ratio and Fe levels in male MDD patients were significantly linked to neurometabolic alterations in the ACC.
Collapse
Affiliation(s)
- Qilin Zhong
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Shunkai Lai
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Jiali He
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Shuming Zhong
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou 510630, China.
| | - Xiaodong Song
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Ying Wang
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Yiliang Zhang
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Guanmao Chen
- Medical Imaging Center, First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Shuya Yan
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital, Jinan University, Guangzhou 510630, China.
| |
Collapse
|
2
|
Shamabadi A, Karimi H, Fallahzadeh MA, Vaseghi S, Arabzadeh Bahri R, Fallahpour B, Abdolghaffari AH, Akhondzadeh S. Sex-controlled differences in sertraline and citalopram efficacies in major depressive disorder: a randomized, double-blind trial. Int Clin Psychopharmacol 2024:00004850-990000000-00136. [PMID: 38640201 DOI: 10.1097/yic.0000000000000550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
To investigate the response to antidepressants while controlling for sex, which has been controversial, 92 outpatient males and females with major depressive disorder were assigned to sertraline (100 mg/day) or citalopram (40 mg/day) in two strata and were assessed using Hamilton depression rating scale (HDRS) scores and brain-derived neurotrophic factor (BDNF), interleukin (IL)-6 and cortisol serum levels in this 8-week, randomized, parallel-group, double-blind clinical trial. Data of 40 sertraline and 40 citalopram recipients with equal representation of males and females assigned to each medication were analyzed, while their baseline characteristics were not statistically different (P > 0.05). There were no significant differences between sertraline and citalopram recipients in outcome changes (P > 0.05), all of which indicated improvement, but a significant time-treatment-sex interaction effect in BDNF levels was observed (P = 0.035). Regarding this, subgroup analyses illustrated a significantly greater increase in male BDNF levels following sertraline treatment (P = 0.020) with a moderate to large effect size (Cohen's d = 0.76 and ). Significant associations were observed between percentage changes in IL-6 levels and BDNF levels in sertraline recipients (P = 0.033) and HDRS scores in citalopram recipients (P < 0.001). Sex was an effect modifier in BDNF alterations following sertraline and citalopram administration. Further large-scale, high-quality, long-term studies are recommended.
Collapse
Affiliation(s)
- Ahmad Shamabadi
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran
| | - Hanie Karimi
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran
| | - Mohammad Ali Fallahzadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran
| | - Salar Vaseghi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj
| | - Razman Arabzadeh Bahri
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran
| | - Bita Fallahpour
- Department of Psychiatry, Razi Hospital, University of Social Welfare and Rehabilitation Sciences
| | | | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran
| |
Collapse
|
3
|
Saad N, Raviv D, Mizrachi Zer-Aviv T, Akirav I. Cannabidiol Modulates Emotional Function and Brain-Derived Neurotrophic Factor Expression in Middle-Aged Female Rats Exposed to Social Isolation. Int J Mol Sci 2023; 24:15492. [PMID: 37895171 PMCID: PMC10607116 DOI: 10.3390/ijms242015492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Aging is associated with changes in cognitive and emotional function. Cannabidiol (CBD) has been reported to attenuate stress and anxiety in human and animal studies. In this study, we aimed to assess the therapeutic potential of CBD among middle-aged female rats exposed to social isolation (SI) and the potential involvement of brain-derived neurotrophic factor (BDNF) in these effects. Thirteen-month-old female rats were group-housed (GH) or exposed to social isolation (SI) and treated with vehicle or CBD (10 mg/kg). CBD restored the SI-induced immobility in the forced swim test and the SI-induced decrease in the expression of BDNF protein levels in the nucleus accumbens (NAc). CBD also increased the time that rats spent in the center in an open field, improved spatial training, and increased BDNF expression in the medial prefrontal cortex (mPFC) and basolateral amygdala (BLA). BDNF expression was found to be correlated with an antidepressant (in the NAc) and an anxiolytic (in the mPFC, BLA, NAc) phenotype, and with learning improvement in the PFC. Together, our results suggest that CBD may serve as a beneficial agent for wellbeing in old age and may help with age-related cognitive decline.
Collapse
Affiliation(s)
- Nadya Saad
- Department of Psychology, School of Psychological Sciences, University of Haifa, Haifa 3498838, Israel; (N.S.); (D.R.); (T.M.Z.-A.)
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Danielle Raviv
- Department of Psychology, School of Psychological Sciences, University of Haifa, Haifa 3498838, Israel; (N.S.); (D.R.); (T.M.Z.-A.)
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Tomer Mizrachi Zer-Aviv
- Department of Psychology, School of Psychological Sciences, University of Haifa, Haifa 3498838, Israel; (N.S.); (D.R.); (T.M.Z.-A.)
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Irit Akirav
- Department of Psychology, School of Psychological Sciences, University of Haifa, Haifa 3498838, Israel; (N.S.); (D.R.); (T.M.Z.-A.)
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
4
|
James KA, Stromin JI, Steenkamp N, Combrinck MI. Understanding the relationships between physiological and psychosocial stress, cortisol and cognition. Front Endocrinol (Lausanne) 2023; 14:1085950. [PMID: 36950689 PMCID: PMC10025564 DOI: 10.3389/fendo.2023.1085950] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/14/2023] [Indexed: 03/08/2023] Open
Abstract
Stress is viewed as a state of real or perceived threat to homeostasis, the management of which involves the endocrine, nervous, and immune systems. These systems work independently and interactively as part of the stress response. The scientific stress literature, which spans both animal and human studies, contains heterogeneous findings about the effects of stress on the brain and the body. This review seeks to summarise and integrate literature on the relationships between these systems, examining particularly the roles of physiological and psychosocial stress, the stress hormone cortisol, as controlled by the hypothalamic-pituitary-adrenal (HPA) axis, and the effects of stress on cognitive functioning. Health conditions related to impaired HPA axis functioning and their associated neuropsychiatric symptoms will also be considered. Lastly, this review will provide suggestions of clinical applicability for endocrinologists who are uniquely placed to measure outcomes related to endocrine, nervous and immune system functioning and identify areas of intervention.
Collapse
Affiliation(s)
- Katharine Ann James
- Applied Cognitive Science and Experimental Neuropsychology Team (ACSENT) Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
- Division of Geriatric Medicine, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Juliet Ilena Stromin
- Applied Cognitive Science and Experimental Neuropsychology Team (ACSENT) Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
| | - Nina Steenkamp
- Applied Cognitive Science and Experimental Neuropsychology Team (ACSENT) Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
| | - Marc Irwin Combrinck
- Division of Geriatric Medicine, Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
5
|
Curtis GR, Gargiulo AT, Carpenter BA, Pirino BE, Hawks A, Coleman SA, Syed NA, Gupta A, Barson JR. Sex-related differences in endogenous pituitary adenylate cyclase-activating polypeptide (PACAP) in the thalamic paraventricular nucleus: Implications for addiction neuroscience. ADDICTION NEUROSCIENCE 2023; 5:100058. [PMID: 36798694 PMCID: PMC9928148 DOI: 10.1016/j.addicn.2022.100058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Males and females exhibit differences in motivated and affective behavior; however, the neural substrates underlying these differences remain poorly understood. In the paraventricular nucleus of the thalamus (PVT), sex-related differences in neuronal activity have been identified in response to motivated behavior tasks and affective challenges. Within the PVT, the neuropeptide, pituitary adenylate cyclase-activating polypeptide (PACAP), is highly expressed and is also involved in motivated and affective behavior. The purpose of this study was to compare the expression of PACAP mRNA and peptide in the PVT of males and females. Analysis with quantitative real-time PCR in mice revealed that females had significantly higher levels of PACAP mRNA than males in the whole PVT, but no differences in the neuropeptides enkephalin or corticotropin releasing factor (CRF) in this brain region. While in rats, females demonstrated a trend for greater gene expression than males in the anterior/middle and middle/posterior PVT, they again showed no differences in enkephalin or CRF. Analysis with immunofluorescent histochemistry revealed that female mice had significantly more PACAP-containing cells than males as a function of area throughout the PVT, and that female rats had significantly more PACAP-27 and PACAP-38-containing cells than males, both as a percentage of total cells and as a function of PVT area. For PACAP-27, this specifically occurred in the anterior PVT, and for PACAP-38, it occurred throughout the anterior, middle, and posterior PVT. These results suggest that sex-related differences in PVT PACAP may underly some of the established sex-related differences in motivated and affective behavior.
Collapse
Affiliation(s)
- Genevieve R. Curtis
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - Andrew T. Gargiulo
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - Brody A. Carpenter
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - Breanne E. Pirino
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - Annie Hawks
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - Sierra A. Coleman
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - Nawal A. Syed
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - Anuranita Gupta
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - Jessica R. Barson
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| |
Collapse
|
6
|
The effect of chronic stress on behaviors, inflammation and lymphocyte subtypes in male and female rats. Behav Brain Res 2023; 439:114220. [PMID: 36414104 DOI: 10.1016/j.bbr.2022.114220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/05/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Excessively released proinflammatory mediators from activated macrophages and lymphocytes may contribute to the etiology of depression. However, the relationship between lymphocytes and depression is not fully understood. Although women have higher depression risk than men, sex/gender differences in psychoneuroimmunological mechanisms are still unclear. To explore these two questions, chronic unpredictable mild stress (CUMS) was used to evaluate the changes in behaviors, inflammation and lymphocyte subtypes in adult male and female Wistar rats. Results show that CUMS increased anhedonia and anxiety-like behaviors, along with increased serum corticosterone, hippocampal pro-inflammatory factors, CD11b, IFN-γ, IL-6 and IL-17, but decreased CD4, CD25, CD4/CD8 ratio, GFAP, 5-hydroxytryptamine (5-HT) and NE concentrations, regardless of sex. There was no positive correlation between sucrose preference and blood CD4/CD8 ratio, but a positive correlation between sucrose preference and spleen CD25, sucrose preference and neurotransmitters (NE and 5-HT), spleen CD25 and serum TGF-β1/IL-6 ratio were found, regardless of sex. Females presented higher basal locomotion, blood CD4, CD4/CD8 ratio, serum corticosteroid and IL-6 concentrations, but lower hippocampal norepinephrine (NE) than males. Although CUMS didn't induce significant sex differences, females presented more changes in CD4 and CD8 lymphocytes than male rats. CUMS caused abnormalities in corticosteroid, lymphocytes, cytokines and neurotransmitters, which might be the precursors for inducing depression-like behaviors in both sexes.
Collapse
|
7
|
Parekh SV, Adams LO, Barkell GA, Lysle DT. Sex-differences in anxiety, neuroinflammatory markers, and enhanced fear learning following chronic heroin withdrawal. Psychopharmacology (Berl) 2023; 240:347-359. [PMID: 36633660 PMCID: PMC9879843 DOI: 10.1007/s00213-023-06310-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/31/2022] [Indexed: 01/13/2023]
Abstract
Post-traumatic stress disorder (PTSD) and opioid use disorder (OUD) are comorbid in clinical populations. However, both pre-clinical and clinical studies of these co-occurring disorders have disproportionately represented male subjects, limiting the applicability of these findings. Our previous work has identified chronic escalating heroin administration and withdrawal can produce enhanced fear learning. This behavior is associated with an increase in dorsal hippocampal (DH) interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and glial fibrillary acidic protein (GFAP) immunoreactivity. Further, we have shown that these increases in IL-1β and TNF-α are mechanistically necessary for the development of enhanced fear learning. Although these are exciting findings, this paradigm has only been studied in males. The current studies aim to examine sex differences in the behavioral and neuroimmune effects of chronic heroin withdrawal and future enhanced fear learning. In turn, we determined that chronic escalating heroin administration can produce withdrawal in female rats comparable to male rats. Subsequently, we examined the consequence of heroin withdrawal on future enhanced fear learning and IL-1β, TNF-α, and GFAP immunoreactivity. Strikingly, we identified sex differences in these neuroimmune measures, as chronic heroin administration and withdrawal does not produce enhanced fear learning or immunoreactivity changes in females. Moreover, we determined whether heroin withdrawal produces short-term and long-term anxiety behaviors in both female and males. Collectively, these novel experiments are the first to test whether heroin withdrawal can sensitize future fear learning, produce neurobiological changes, and cause short-term and long-term anxiety behaviors in female rats.
Collapse
Affiliation(s)
- Shveta V Parekh
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA
| | - Lydia O Adams
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA
| | - Gillian A Barkell
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA
| | - Donald T Lysle
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA.
| |
Collapse
|
8
|
Helman TJ, Headrick JP, Stapelberg NJC, Braidy N. The sex-dependent response to psychosocial stress and ischaemic heart disease. Front Cardiovasc Med 2023; 10:1072042. [PMID: 37153459 PMCID: PMC10160413 DOI: 10.3389/fcvm.2023.1072042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Stress is an important risk factor for modern chronic diseases, with distinct influences in males and females. The sex specificity of the mammalian stress response contributes to the sex-dependent development and impacts of coronary artery disease (CAD). Compared to men, women appear to have greater susceptibility to chronic forms of psychosocial stress, extending beyond an increased incidence of mood disorders to include a 2- to 4-fold higher risk of stress-dependent myocardial infarction in women, and up to 10-fold higher risk of Takotsubo syndrome-a stress-dependent coronary-myocardial disorder most prevalent in post-menopausal women. Sex differences arise at all levels of the stress response: from initial perception of stress to behavioural, cognitive, and affective responses and longer-term disease outcomes. These fundamental differences involve interactions between chromosomal and gonadal determinants, (mal)adaptive epigenetic modulation across the lifespan (particularly in early life), and the extrinsic influences of socio-cultural, economic, and environmental factors. Pre-clinical investigations of biological mechanisms support distinct early life programming and a heightened corticolimbic-noradrenaline-neuroinflammatory reactivity in females vs. males, among implicated determinants of the chronic stress response. Unravelling the intrinsic molecular, cellular and systems biological basis of these differences, and their interactions with external lifestyle/socio-cultural determinants, can guide preventative and therapeutic strategies to better target coronary heart disease in a tailored sex-specific manner.
Collapse
Affiliation(s)
- Tessa J. Helman
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
- Correspondence: Tessa J. Helman
| | - John P. Headrick
- Schoolof Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | | | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
| |
Collapse
|
9
|
Prior short-term exercise prevents behavioral and biochemical abnormalities induced by single prolonged stress in a rat model of posttraumatic stress disorder. Behav Brain Res 2022; 428:113864. [DOI: 10.1016/j.bbr.2022.113864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/27/2022] [Accepted: 03/27/2022] [Indexed: 11/21/2022]
|
10
|
Olave FA, Aguayo FI, Román-Albasini L, Corrales WA, Silva JP, González PI, Lagos S, García MA, Alarcón-Mardones M, Rojas PS, Xu X, Cidlowski JA, Aliaga E, Fiedler J. Chronic restraint stress produces sex-specific behavioral and molecular outcomes in the dorsal and ventral rat hippocampus. Neurobiol Stress 2022; 17:100440. [PMID: 35252485 PMCID: PMC8894263 DOI: 10.1016/j.ynstr.2022.100440] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/09/2022] [Accepted: 02/21/2022] [Indexed: 01/24/2023] Open
Abstract
Stress-related disorders display differences at multiple levels according to sex. While most studies have been conducted in male rodents, less is known about comparable outcomes in females. In this study, we found that the chronic restraint stress model (2.5 h/day for 14 days) triggers different somatic responses in male and female adult rats. Chronic restraint produced a loss in sucrose preference and novel location preference in male rats. However, chronic restraint failed to produce loss of sucrose preference in females, while it improved spatial performance. We then characterized the molecular responses associated with these behaviors in the hippocampus, comparing the dorsal and ventral poles. Notably, sex- and hippocampal pole-specific transcriptional signatures were observed, along with a significant concordance between the female ventral and male dorsal profiles. Functional enrichment analysis revealed both shared and specific terms associated with each pole and sex. By looking into signaling pathways that were associated with these terms, we found an ample array of sex differences in the dorsal and, to a lesser extent, in the ventral hippocampus. These differences were mainly present in synaptic TrkB signaling, Akt pathway, and glutamatergic receptors. Unexpectedly, the effects of stress on these pathways were rather minimal and mostly dissociated from the sex-specific behavioral outcomes. Our study suggests that female rats are resilient and males susceptible to the restraint stress exposure in the sucrose preference and object location tests, while the activity of canonical signaling pathways is primarily determined by sex rather than stress in the dorsal and ventral hippocampus.
Collapse
Affiliation(s)
- Felipe A. Olave
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Felipe I. Aguayo
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Luciano Román-Albasini
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Wladimir A. Corrales
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Juan P. Silva
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Pablo I. González
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Sara Lagos
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - María A. García
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Matías Alarcón-Mardones
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Paulina S. Rojas
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Xiaojiang Xu
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, 27709, USA
| | - John A. Cidlowski
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, 27709, USA
| | - Esteban Aliaga
- Medical Technology School and the Neuropsychology and Cognitive Neurosciences Research Center (CINPSI-Neurocog), Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile
- Corresponding author. Medical Technology School, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile.
| | - Jenny Fiedler
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
- Corresponding author. Department of Biochemistry and Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago. Chile.
| |
Collapse
|
11
|
Mujinya R, Kalange M, Ochieng JJ, Ninsiima HI, Eze ED, Afodun AM, Nabirumbi R, Sulaiman SO, Kairania E, Echoru I, Okpanachi AO, Matama K, Asiimwe OH, Nambuya G, Usman IM, Obado OL, Zirintunda G, Ssempijja F, Nansunga M, Matovu H, Ayikobua ET, Nganda PE, Onanyang D, Ekou J, Musinguzi SP, Ssimbwa G, Kasozi KI. Cerebral Cortical Activity During Academic Stress Amongst Undergraduate Medical Students at Kampala International University (Uganda). Front Psychiatry 2022; 13:551508. [PMID: 35757206 PMCID: PMC9231459 DOI: 10.3389/fpsyt.2022.551508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 03/29/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Stress among medical students is related to their academic lifespan; however, information on brain health among medical students from developing countries continues to be scarce. The objective of this study was to establish perceived academic stress levels, assess the ability to cope with stress, and investigate its effects on the visual reaction time (VRT), audio reaction time (ART), and tactile reaction time (TRT) in the somatosensory cortex among medical students of Uganda. METHODS This was a cross-sectional study conducted among preclinical (n = 88) and clinical (n = 96) undergraduate medical students at Kampala International University Western Campus. A standard Perceived Stress Scale (PSS) was used to categorize stress into low, moderate, and severe while the ability to cope with stress was categorized into below average, average, above average, and superior stresscoper (SS). Data on reaction time were acquired through VRT, ART, and TRT using the catch-a-ruler experiment, and this was analyzed using SPSS version 20. RESULTS This study shows that preclinical students are more stressed than clinical students (PSS prevalence for low stress = preclinical; clinical: 40, 60%). Moderate stress was 48.4 and 51.6% while high perceived stress was 75 and 25% among preclinical and clinical students. Among male and female students in preclinical years, higher TRT and VRT were found in clinical students showing that stress affects the tactile and visual cortical areas in the brain, although the VRT scores were only significantly (P = 0.0123) poor in male students than female students in biomedical sciences. Also, highly stressed individuals had higher TRT and ART and low VRT. SS had high VRT and ART and low TRT in preclinical students, demonstrating the importance of the visual cortex in stress plasticity. Multiple regression showed a close relationship between PSS, ability to cope with stress, age, and educational level (P < 0.05), demonstrating the importance of social and psychological support, especially in the biomedical sciences. CONCLUSION Preclinical students suffer more from stress and are poorer SS than clinical students. This strongly impairs their cortical regions in the brain, thus affecting their academic productivity.
Collapse
Affiliation(s)
- Regan Mujinya
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Muhamudu Kalange
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Juma John Ochieng
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | | | | | - Adam Moyosore Afodun
- Department of Anatomy and Cell Biology, Faculty of Health Sciences, Busitema University, Tororo, Uganda
| | | | - Sheu Oluwadare Sulaiman
- Graduate Program in Cell Biology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Emmanuel Kairania
- Department of Anatomy and Cell Biology, Faculty of Health Sciences, Busitema University, Tororo, Uganda
| | - Isaac Echoru
- School of Medicine, Kabale University, Kabale, Uganda
| | | | - Kevin Matama
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy, Kampala International University Western Campus, Bushenyi, Uganda
| | - Oscar Hilary Asiimwe
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Grace Nambuya
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Ibe Michael Usman
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | | | - Gerald Zirintunda
- Department of Animal Production and Management, Faculty of Agriculture and Animal Sciences, Busitema University, Tororo, Uganda
| | - Fred Ssempijja
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Miriam Nansunga
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda.,Department of Physiology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Henry Matovu
- Department of Animal Production and Management, Faculty of Agriculture and Animal Sciences, Busitema University, Tororo, Uganda
| | | | - Ponsiano Ernest Nganda
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - David Onanyang
- Department of Biology, Faculty of Science, Gulu University, Gulu, Uganda
| | - Justine Ekou
- Department of Animal Production and Management, Faculty of Agriculture and Animal Sciences, Busitema University, Tororo, Uganda
| | - Simon Peter Musinguzi
- Department of Agriculture Production, Faculty of Agriculture, Kyambogo University, Kampala, Uganda
| | - Godfrey Ssimbwa
- Department of Physiology, Faculty of Health Sciences, Muni University, Arua, Uganda
| | | |
Collapse
|
12
|
Wei F, Zhang L, Ma B, Li W, Deng X, Zheng T, Wang X, Jing Y. Oxytocin system driven by experiences modifies social recognition and neuron morphology in female BALB/c mice. Peptides 2021; 146:170659. [PMID: 34571057 DOI: 10.1016/j.peptides.2021.170659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 11/20/2022]
Abstract
The oxytocin (OT) system, affected by life experiences, modulates neuron morphology in a sex-specific manner, leading to sex differences in social interactions. To date, few studies have focused on the OT system and social interactions of female mice. In this study, we used maternal deprivation (MD) and its possible treatment, environmental enrichment (EE), to affect social recognition in female BALB/c mice. We checked neuron morphology, synaptic connections, oxytocinergic (OTergic) neurons in the hypothalamus paraventricular nucleus (PVH), and OT receptor (OTR) in the basolateral amygdala (BLA) and layer II/III of the prelimbic cortex (PL). Our results showed that MD induced social recognition impairments, increased OTR levels in the BLA, and, meanwhile, reduced OTergic neurons in the magnocellular region of the PVH (mPVH). Decreased Nissl bodies, increased cell nuclei, and increased dendrites of projection neurons paralleled the increased OTR levels in the BLA of MD mice. EE restored MD-induced the impairments of novel object recognition and sociability; this effect paralleled a decrease in cell density in the PL and an increase in OTergic neurons in the parvocellular regions of the PVH and synaptic connections in the BLA and layer II/III of the PL. Our findings indicate that early life stress such as MD impairs social recognition, and meanwhile, remodels neuron morphology region-specifically in the female brain, apparently in the BLA but slightly in the PL; and EE could partially restore the deficits induced by MD. The results provide new insights into sex differences in the prevalence of psychological development disorders.
Collapse
Affiliation(s)
- Fengmei Wei
- Department of Physiology and Psychology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, 730000, PR China; Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Lang Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Bo Ma
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Wenhao Li
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Xiao Deng
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Tingjuan Zheng
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, PR China
| | - Xiaohui Wang
- Department of Nuclear Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu Province, 730000, PR China
| | - Yuhong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu, 730000, PR China.
| |
Collapse
|
13
|
El Marzouki H, Aboussaleh Y, Najimi M, Chigr F, Ahami A. Effect of Cold Stress on Neurobehavioral and Physiological Parameters in Rats. Front Physiol 2021; 12:660124. [PMID: 34603068 PMCID: PMC8485037 DOI: 10.3389/fphys.2021.660124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Cold stress is an important current issue and implementing control strategies to limit its sometimes harmful effects is crucial. Cold is a common stressor that can occur in our work and our occupational or leisure time activities every day. There are substantial studies on the effects of chronic stress on memory and behavior, although, the cognitive changes and anxiety disorders that can occur after exposure to chronic intermittent cold stress are not completely characterized. Therefore, the present study was undertaken with an aim to investigate the effects of chronic intermittent cold stress on body weight, food intake and working memory, and to elucidate cold stress related anxiety disorders using cognitive and behavioral test batteries. Methods: We generated a cold stress model by exposing rats to chronic intermittent cold stress for 5 consecutive days and in order to test for the potential presence of sex differences, a comparable number of male and female rats were tested in the current study. Then, we measured the body weights, food intake and the adrenal glands weight. Working memory and recognition memory were assessed using the Y maze and the Novel Object Recognition (NOR) tasks. While, sex differences in the effects of chronic stress on behavior were evaluated by the elevated plus maze (EPM), open field maze (OF), and Marble burying (MB) tests. Results: We found that 2 h exposure to cold (4°C) resulted in an increase in the relative weight of the adrenal glands in male rats. Given the same chronic stress 5 days of cold exposure (2 h per day), increased weight gain in male rats, while females showed decreased food intake and no change in body weight. Both sexes successfully performed the Y maze and object recognition (OR) tasks, indicating intact spatial working memory performance and object recognition abilities in both male and female rats. In addition, we have shown that stress caused an increase in the level of anxiety in male rats. In contrast, the behavior of the female rats was not affected by cold exposure. Conclusion: Overall, the current results provide preliminary evidence that chronic intermittent cold stress model may not be an efficient stressor to female rats. Females exhibit resilience to cold exposure that causes an increase in the level of anxiety in male rats, which demonstrates that they are affected differently by stress and the gender is an important consideration in experimental design.
Collapse
Affiliation(s)
- Hajar El Marzouki
- Biology and Health Laboratory, Unit of Clinical and Cognitive-Behavioural Neurosciences and Applied Nutrition Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Youssef Aboussaleh
- Biology and Health Laboratory, Unit of Clinical and Cognitive-Behavioural Neurosciences and Applied Nutrition Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Mohamed Najimi
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan MoulaySlimane University, Beni Mellal, Morocco
| | - Fatiha Chigr
- Biological Engineering Laboratory, Faculty of Sciences and Techniques, Sultan MoulaySlimane University, Beni Mellal, Morocco
| | - Ahmed Ahami
- Biology and Health Laboratory, Unit of Clinical and Cognitive-Behavioural Neurosciences and Applied Nutrition Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| |
Collapse
|
14
|
Du Preez A, Eum J, Eiben I, Eiben P, Zunszain PA, Pariante CM, Thuret S, Fernandes C. Do different types of stress differentially alter behavioural and neurobiological outcomes associated with depression in rodent models? A systematic review. Front Neuroendocrinol 2021; 61:100896. [PMID: 33359461 DOI: 10.1016/j.yfrne.2020.100896] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022]
Abstract
Cataloguing the effects of different types of stress on behaviour and physiology in rodent models has not been comprehensively attempted. Here, we systematically review whether chronic exposure to physical stress, psychosocial stress, or both types of stress can induce different behavioural and neurobiological outcomes in male and female rodents. We found that physical stress consistently increased depressive-like behaviour, impaired social interaction and decreased body weight, while psychosocial stress consistently increased both anxiety- and depressive-like behaviour, impaired social interaction and learning and memory, increased HPA axis activity, peripheral inflammation and microglial activation, and decreased hippocampal neurogenesis in male rodents. Moreover, we found that the combined effect of both stress types resulted in a more severe pathological state defined by increased anxiety- and depressive-like behaviour, impaired social interaction and learning and memory, increased HPA axis activity and central inflammation, and reduced hippocampal neurogenesis and neural plasticity in male rodents. Phenotypes for females were less consistent, irrespective of the type of stress exposure, on account of the limited number of studies using females. This review highlights that the type of stress may indeed matter and will help animal researchers to more appropriately choose a stress/depression model that fits their research purposes.
Collapse
Affiliation(s)
- Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK.
| | - Josephine Eum
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Inez Eiben
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Paola Eiben
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Patricia A Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK
| |
Collapse
|
15
|
Selecting antidepressants according to a drug-by-environment interaction: A comparison of fluoxetine and minocycline effects in mice living either in enriched or stressful conditions. Behav Brain Res 2021; 408:113256. [PMID: 33775780 DOI: 10.1016/j.bbr.2021.113256] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/18/2021] [Accepted: 03/18/2021] [Indexed: 11/23/2022]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the first-line treatment for major depressive disorder. It has been recently proposed that these drugs, by enhancing neural plasticity, amplify the influences of the living conditions on mood. Consequently, SSRI outcome depends on the quality of the environment, improving symptomatology mainly in individuals living in favorable conditions. In adverse conditions, drugs with a different mechanism of action might have higher efficacy. The antibiotic minocycline, with neuroprotective and anti-inflammatory properties, has been recently proposed as a novel potential antidepressant treatment. To explore the drug-by-environment interaction, we compared the effects on depressive-like behavior and neural plasticity of the SSRI fluoxetine and minocycline in enriched and stressful conditions. We first exposed C57BL/6 adult female mice to 14 days of chronic unpredictable mild stress to induce a depressive-like profile. Afterward, mice received vehicle, fluoxetine, or minocycline for 21 days, while exposed to either enriched or stressful conditions. During the first five days, fluoxetine led to an improvement in enrichment but not in stress. By contrast, minocycline led to an improvement in both conditions. After 21 days, all groups showed a significant improvement in enrichment while fluoxetine worsened the depressive like behavior in stress. The effects of the drugs on neural plasticity, measured as long-term potentiation, were also environment-dependent. Overall, we show that the environment affects fluoxetine but not minocycline outcome, indicating that the latter represents a potential alternative to SSRIs to treat depressed patients living in adverse conditions. From a translation perspective, our finding call for considering the drug-by-environment interaction to select the most effective pharmacological treatment.
Collapse
|
16
|
Alshammari TK. Sexual dimorphism in pre-clinical studies of depression. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110120. [PMID: 33002519 DOI: 10.1016/j.pnpbp.2020.110120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022]
Abstract
Although there is a sex bias in the pathological mechanisms exhibited by brain disorders, investigation of the female brain in biomedical science has long been neglected. Use of the male model has generally been the preferred option as the female animal model exhibits both biological variability and hormonal fluctuations. Existing studies that compare behavioral and/or molecular alterations in animal models of brain diseases are generally underrepresented, and most utilize the male model. Nevertheless, in recent years there has been a trend toward the increased inclusion of females in brain studies. However, current knowledge regarding sex-based differences in depression and stress-related disorders is limited. This can be improved by reviewing preclinical studies that highlight sex differences in depression. This paper therefore presents a review of sex-based preclinical studies of depression. These shed light on the discrepancies between males and females regarding the biological mechanisms that underpin mechanistic alterations in the diseased brain. This review also highlights the conclusions drawn by preclinical studies to advance our understanding of mood disorders, encouraging researchers to promote ways of investigating and managing sexually dimorphic disorders.
Collapse
Affiliation(s)
- Tahani K Alshammari
- Department of Pharmacology and Toxicology, Pharmacy College, King Saud University, Saudi Arabia; Prince Naïf Bin Abdul-Aziz Health Research Center, King Saud University, Saudi Arabia.
| |
Collapse
|
17
|
Gupta K, Chattarji S. Sex differences in the delayed impact of acute stress on the amygdala. Neurobiol Stress 2021; 14:100292. [PMID: 33490316 PMCID: PMC7807162 DOI: 10.1016/j.ynstr.2020.100292] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/10/2020] [Accepted: 12/28/2020] [Indexed: 01/02/2023] Open
Abstract
There is accumulating evidence that stress triggers specific temporal patterns of morphological plasticity in the amygdala, a brain area that plays a pivotal role in the debilitating emotional symptoms of stress-related psychiatric disorders. Acute immobilization stress is known to cause a delayed increase in the density of dendritic spines on principal neurons in the basolateral amygdala (BLA) of rats. These neuronal changes are also accompanied by a delayed enhancement in anxiety-like behavior. However, these earlier studies used male rats, and the delayed behavioral and synaptic effects of acute stress on the BLA of female rats remain unexplored. Here, using whole-cell recordings in rat brain slices, we find that a single exposure to 2-h immobilization stress leads to an increase, 10 days later, in the frequency of miniature excitatory postsynaptic currents (mEPSCs) recorded from lateral amygdala (LA) principal neurons in male rats. Further, acute stress also causes a reduction in the frequency of miniature inhibitory postsynaptic currents (mIPSCs) in LA neurons 10 days after acute stress. In striking contrast, excitatory and inhibitory synaptic transmission in the LA of female rats does not exhibit any delayed change despite exposure to the same acute stress. Finally, we examined the functional impact of these contrasting synaptic changes at the behavioral level. Male rats exhibit a delayed increase in anxiety-like behavior on the elevated plus-maze 10 days after acute stress. However, the same stress does not lead to a delayed anxiogenic effect in female rats. Together, these results demonstrate that the delayed modulation of the balance of synaptic excitation and inhibition in the amygdala, as well as anxiety-like behavior, differ between males and females. These findings provide a framework, across biological scales, for analyzing how affective symptoms of stress disorders vary between the sexes.
Collapse
Affiliation(s)
- Kanika Gupta
- National Centre for Biological Sciences, Bangalore, 560065, India
| | | |
Collapse
|
18
|
Faraji J, Metz GAS. Infrared Thermography Reveals Sex-Specific Responses to Stress in Mice. Front Behav Neurosci 2020; 14:79. [PMID: 32523518 PMCID: PMC7261839 DOI: 10.3389/fnbeh.2020.00079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/28/2020] [Indexed: 01/20/2023] Open
Abstract
Psychogenic hyperthermia is a stress-related condition reported mostly in women. Neuroendocrine responses to stress in females differ from those in males, and these differences cannot be explained solely based on hypothalamic-pituitary-adrenal (HPA) axis activity. Here, we used infrared (IR) thermographic imaging to record changes in cutaneous temperature following two types of stressful experiences in female and male mice. Mice were exposed to either single-session restraint stress or vertical exploration (rearing) deprivation and were monitored for exploratory activity and IR surface thermal changes. Females displayed higher rearing activity than males during the dark phase of the light cycle. Both sexes showed similar plasma corticosterone (CORT) responses after a challenge with restraint and rearing deprivation. However, only females responded to rearing deprivation with increased cutaneous temperature in the head and back, and a reduced thermal response in the tail. Circulating CORT levels were not correlated with the thermal variations. These findings, for the first time, provide evidence for sex-specific cutaneous thermal responses to short-term stress in mice following transient vertical-activity deprivation that may mimic clinical psychogenic hyperthermia.
Collapse
Affiliation(s)
- Jamshid Faraji
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada.,Faculty of Nursing & Midwifery, Golestan University of Medical Sciences, Gorgan, Iran
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
19
|
Perrino C, Ferdinandy P, Bøtker HE, Brundel BJJM, Collins P, Davidson SM, den Ruijter HM, Engel FB, Gerdts E, Girao H, Gyöngyösi M, Hausenloy DJ, Lecour S, Madonna R, Marber M, Murphy E, Pesce M, Regitz-Zagrosek V, Sluijter JPG, Steffens S, Gollmann-Tepeköylü C, Van Laake LW, Van Linthout S, Schulz R, Ytrehus K. Improving translational research in sex-specific effects of comorbidities and risk factors in ischaemic heart disease and cardioprotection: position paper and recommendations of the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2020; 117:367-385. [PMID: 32484892 DOI: 10.1093/cvr/cvaa155] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/29/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Ischaemic heart disease (IHD) is a complex disorder and a leading cause of death and morbidity in both men and women. Sex, however, affects several aspects of IHD, including pathophysiology, incidence, clinical presentation, diagnosis as well as treatment and outcome. Several diseases or risk factors frequently associated with IHD can modify cellular signalling cascades, thus affecting ischaemia/reperfusion injury as well as responses to cardioprotective interventions. Importantly, the prevalence and impact of risk factors and several comorbidities differ between males and females, and their effects on IHD development and prognosis might differ according to sex. The cellular and molecular mechanisms underlying these differences are still poorly understood, and their identification might have important translational implications in the prediction or prevention of risk of IHD in men and women. Despite this, most experimental studies on IHD are still undertaken in animal models in the absence of risk factors and comorbidities, and assessment of potential sex-specific differences are largely missing. This ESC WG Position Paper will discuss: (i) the importance of sex as a biological variable in cardiovascular research, (ii) major biological mechanisms underlying sex-related differences relevant to IHD risk factors and comorbidities, (iii) prospects and pitfalls of preclinical models to investigate these associations, and finally (iv) will provide recommendations to guide future research. Although gender differences also affect IHD risk in the clinical setting, they will not be discussed in detail here.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary.,Pharmahungary Group, Hajnoczy str. 6., H-6722 Szeged, Hungary
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 161, 8200 Aarhus, Denmark
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, Amsterdam, 1108 HV, the Netherlands
| | - Peter Collins
- Imperial College, Faculty of Medicine, National Heart & Lung Institute, South Kensington Campus, London SW7 2AZ, UK.,Royal Brompton Hospital, Sydney St, Chelsea, London SW3 6NP, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Hester M den Ruijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), Schwabachanlage 12, 91054 Erlangen, Germany
| | - Eva Gerdts
- Department for Clinical Science, University of Bergen, PO Box 7804, 5020 Bergen, Norway
| | - Henrique Girao
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, and Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, 169609, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, 119228, Singapore.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, Chris Barnard Building, University of Cape Town, Private Bag X3 7935 Observatory, Cape Town, South Africa
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy.,Department of Internal Medicine, University of Texas Medical School in Houston, 6410 Fannin St #1014, Houston, TX 77030, USA
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Elizabeth Murphy
- Laboratory of Cardiac Physiology, Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD 20892, USA
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS Via Parea, 4, I-20138 Milan, Italy
| | - Vera Regitz-Zagrosek
- Berlin Institute of Gender in Medicine, Center for Cardiovascular Research, DZHK, partner site Berlin, Geschäftsstelle Potsdamer Str. 58, 10785 Berlin, Germany.,University of Zürich, Rämistrasse 71, 8006 Zürich, Germany
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands.,Circulatory Health Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Can Gollmann-Tepeköylü
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstr.35, A - 6020 Innsbruck, Austria
| | - Linda W Van Laake
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Ludwigstraße 23, 35390 Giessen, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9037 Tromsø, Norway
| |
Collapse
|
20
|
Miao Z, Wang Y, Sun Z. The Relationships Between Stress, Mental Disorders, and Epigenetic Regulation of BDNF. Int J Mol Sci 2020; 21:ijms21041375. [PMID: 32085670 PMCID: PMC7073021 DOI: 10.3390/ijms21041375] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/01/2020] [Accepted: 02/15/2020] [Indexed: 12/25/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a critical member of the neurotrophic family, plays an important role in multiple stress-related mental disorders. Although alterations in BDNF in multiple brain regions of individuals experiencing stress have been demonstrated in previous studies, it appears that a set of elements are involved in the complex regulation. In this review, we summarize the specific brain regions with altered BDNF expression during stress exposure. How various environmental factors, including both physical and psychological stress, affect the expression of BDNF in specific brain regions are further summarized. Moreover, epigenetic regulation of BDNF, including DNA methylation, histone modification, and noncoding RNA, in response to diverse types of stress, as well as sex differences in the sensitivity of BDNF to the stress response, is also summarized. Clarification of the underlying role of BDNF in the stress process will promote our understanding of the pathology of stress-linked mental disorders and provide a potent target for the future treatment of stress-related illness.
Collapse
Affiliation(s)
- Zhuang Miao
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China;
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China;
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yan Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China;
- School of Life Sciences, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongsheng Sun
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China;
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China;
- School of Life Sciences, University of the Chinese Academy of Sciences, Beijing 100049, China
- Correspondence:
| |
Collapse
|
21
|
Rosinger ZJ, De Guzman RM, Jacobskind JS, Saglimbeni B, Malone M, Fico D, Justice NJ, Forni PE, Zuloaga DG. Sex-dependent effects of chronic variable stress on discrete corticotropin-releasing factor receptor 1 cell populations. Physiol Behav 2020; 219:112847. [PMID: 32081812 DOI: 10.1016/j.physbeh.2020.112847] [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: 11/15/2019] [Revised: 01/25/2020] [Accepted: 02/13/2020] [Indexed: 12/19/2022]
Abstract
Anxiety and depression are strikingly more prevalent in women compared with men. Dysregulation of corticotropin-releasing factor (CRF) binding to its cognate receptor (CRFR1) is thought to play a critical role in the etiology of these disorders. In the present study, we investigated whether there were sex differences in the effects of chronic variable stress (CVS) on CRFR1 cells using CRFR1-GFP reporter mice experiencing a 9-day CVS paradigm. Brains were collected from CVS and stress naïve female and male mice following exposure to the open field test. This CVS paradigm effectively increased anxiety-like behavior in female and male mice. In addition, we assessed changes in activation of CRFR1 cells (co-localization with c-Fos and phosphorylated CREB (pCREB)) in stress associated brain structures, including two sexually dimorphic CRFR1 cell groups in the anteroventral periventricular nucleus (AVPV/PeN; F>M) and paraventricular hypothalamus (PVN; M>F). CVS increased CRFR1-GFP cell number as well as the number of CRFR1/pCREB co-expressing cells in the female but not male AVPV/PeN. In the PVN, the number of CRFR1/pCREB co-expressing cells was overall greater in males regardless of treatment and CVS resulted in a male-specific reduction of CRFR1/c-Fos cells. In addition, CVS induced a female-specific reduction in CRFR1/c-Fos cells within the anteroventral bed nucleus of the stria terminalis and both sexes exhibited a reduction in CRFR1/c-Fos co-expressing cells following CVS within the ventral basolateral amygdala. Overall, these sex-specific effects of CVS on CRFR1 populations may have implications for sex differences in stress-induction of mood disorders.
Collapse
Affiliation(s)
- Zachary J Rosinger
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Rose M De Guzman
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Jason S Jacobskind
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Brianna Saglimbeni
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Margaret Malone
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Danielle Fico
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Nicholas J Justice
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, TX, United States
| | - Paolo E Forni
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, United States
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, Albany, NY 12222, United States.
| |
Collapse
|
22
|
Effects of stress on the structure and function of the medial prefrontal cortex: Insights from animal models. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 150:129-153. [PMID: 32204829 DOI: 10.1016/bs.irn.2019.11.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Stress alters both cognitive and emotional function, and increases risk for a variety of psychological disorders, such as depression and posttraumatic stress disorder. The prefrontal cortex is critical for executive function and emotion regulation, is a target for stress hormones, and is implicated in many stress-influenced psychological disorders. Therefore, understanding how stress-induced changes in the structure and function of the prefrontal cortex are related to stress-induced changes in behavior may elucidate some of the mechanisms contributing to stress-sensitive disorders. This review focuses on data from rodent models to describe the effects of chronic stress on behaviors mediated by the medial prefrontal cortex, the effects of chronic stress on the morphology and physiology of the medial prefrontal cortex, mechanisms that may mediate these effects, and evidence for sex differences in the effects of stress on the prefrontal cortex. Understanding how stress influences prefrontal cortex and behaviors mediated by it, as well as sex differences in this effect, will elucidate potential avenues for novel interventions for stress-sensitive disorders characterized by deficits in executive function and emotion regulation.
Collapse
|
23
|
Blume SR, Padival M, Urban JH, Rosenkranz JA. Disruptive effects of repeated stress on basolateral amygdala neurons and fear behavior across the estrous cycle in rats. Sci Rep 2019; 9:12292. [PMID: 31444385 PMCID: PMC6707149 DOI: 10.1038/s41598-019-48683-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/08/2019] [Indexed: 02/02/2023] Open
Abstract
Stress is a precipitating factor in depression and anxiety disorders. Patients with these disorders often show amygdala abnormalities. The basolateral amygdala (BLA) is integral in mood and emotion, and is sensitive to stress. While much is known about effects of stress on BLA neuron activity and morphology in males, less is known in females. We tested whether repeated stress exerts distinct effects on BLA in vivo neuronal activity and morphology of Golgi-stained BLA neurons [lateral (LAT) and basal (BA) nuclei] in adult female rats. Repeated restraint stress increased BLA neuronal firing and caused hypertrophy of BLA neurons in males, while it decreased LAT and BA neuronal firing and caused hypotrophy of neurons in the LAT of females. BLA neuronal activity and function, such as fear conditioning, shifts across the estrous cycle. Repeated stress disrupted this pattern of BLA activity and fear expression over the estrous cycle. The disruptive effects of stress on the pattern of BLA function across estrous may produce behavior that is non-optimal for a specific phase of the estrous cycle. The contrasting effects of stress may contribute to sex differences in the effects of stress on mood and psychiatric disorders.
Collapse
Affiliation(s)
- Shannon R Blume
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 60064, USA
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL, 60064, USA
| | - Mallika Padival
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 60064, USA
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 60064, USA
| | - Janice H Urban
- Discipline of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 60064, USA
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 60064, USA
| | - J Amiel Rosenkranz
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 60064, USA.
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 60064, USA.
| |
Collapse
|
24
|
Balkaya M, Seidel JL, Sadeghian H, Qin T, Chung DY, Eikermann-Haerter K, van den Maagdenberg AMJM, Ferrari MD, Ayata C. Relief Following Chronic Stress Augments Spreading Depolarization Susceptibility in Familial Hemiplegic Migraine Mice. Neuroscience 2019; 415:1-9. [PMID: 31299346 DOI: 10.1016/j.neuroscience.2019.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 06/29/2019] [Accepted: 07/01/2019] [Indexed: 11/25/2022]
Abstract
Cortical spreading depolarization (CSD) is the electrophysiological substrate of migraine aura, and a putative trigger of trigeminovascular activation and migraine headache. Many migraineurs report stress or relief after a stress triggers an attack. We tested whether various stress conditions might modulate CSD susceptibility and whether this is dependent on genetic factors. Male and female wild type and familial hemiplegic migraine type1 (FHM1) knock-in mice heterozygous for the S218L missense mutation were subjected to acute or chronic stress, or chronic stress followed by relief (36 h). Acute stress was induced by restraint and exposure to bright light and white noise (3 h). Chronic stress was induced for 28 days by two cycles of repeated exposure of mice to a rat (7 days), physical restraint (3 days), and forced swimming (3 days). Electrical CSD threshold and KCl-induced (300 mM) CSD frequency were determined in occipital cortex in vivo at the end of each protocol. Relief after chronic stress reduced the electrical CSD threshold and increased the frequency of KCl-induced CSDs in FHM1 mutants only. Acute or chronic stress without relief did not affect CSD susceptibility in either strain. Stress status did not affect CSD propagation speed, duration or amplitude. In summary, relief after chronic stress, but not acute or chronic stress alone, augments CSD in genetically susceptible mice. Therefore, enhanced CSD susceptibility may explain why, in certain patients, migraine attacks typically occur during a period of stress relief such as weekends or holidays.
Collapse
Affiliation(s)
- Mustafa Balkaya
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jessica L Seidel
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Homa Sadeghian
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Tao Qin
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - David Y Chung
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Katharina Eikermann-Haerter
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Arn M J M van den Maagdenberg
- Department of Neurology Leiden University Medical Center, Leiden 2300, RC, the Netherlands; Human Genetics, Leiden University Medical Center, Leiden 2300, RC, the Netherlands
| | - Michel D Ferrari
- Department of Neurology Leiden University Medical Center, Leiden 2300, RC, the Netherlands
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
25
|
Salama OA, Attia MM, Abdelrazek MAS. Modulatory effects of swimming exercise against malathion induced neurotoxicity in male and female rats. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2019; 157:13-18. [PMID: 31153460 DOI: 10.1016/j.pestbp.2019.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/11/2018] [Accepted: 01/28/2019] [Indexed: 06/09/2023]
Abstract
UNLABELLED Malathion is one of the most commonly used organophosphorus (OP) pesticides. It is important to regard that exposure to OP poisoning may cause anxiety and depression. Malathion toxicity induces cholinergic symptoms. Brain-derived neurotrophic factor (BDNF) is the most profusely expressed neurotrophin in the central nervous system; it promotes the survival of neurons. Regular exercise improves brain well-being and enhances recovery from brain Injuries. It is suggested that BDNF may mediate these effects. Therefore, this study was planned to assess the modulatory effects of regular exercise performance on brain BDNF level, cholinergic activity, oxidative stress and apoptosis in male and female rats subjected to neurotoxicity induced by malathion administration. MATERIALS AND METHODS Thirty-two adult male and thirty-two adult female albino rats were included in this study. The rats were divided into four equal groups (8rats). Control group, malathion treated group, exercised group, malathion exercised group. Acetylcholinesterase (AchE) activity, total antioxidant capacity (TAC), BDNF level and Caspase 3 activity were assessed. RESULTS Female rats had higher baseline content of BDNF in brain homogenate than male rats. Malathion administration induced a significant decrease in BDNF level in female rats and in the total antioxidant capacity in both male and female rats. A significant elevation in caspase 3 activity was detected in the malathion treated groups, with more elevation in female rats. Swimming exercise improved BDNF level, AchE activity, and apoptosis in both male and female rats in all groups. In addition, male rats were more cholinergic system responders to regular exercise than female rats. CONCLUSION It could be concluded that malathion induced elevation in oxidative stress and apoptosis in all rats, with reduction in BDNF level in female rats. Meanwhile, regular swimming exercise was found to improve brain health through modulation of BDNF level and cholinergic activity. It is recommended to practice regular exercise to maintain brain health. Further studies are required to clarify the involvement of sex hormones in BDNF regulation.
Collapse
Affiliation(s)
- Ola A Salama
- Department of Physiology, Medical Research Institute, Alexandria University, Egypt
| | - Maha M Attia
- Department of Physiology, Medical Research Institute, Alexandria University, Egypt
| | - Mohamed A S Abdelrazek
- Department of Chemistry and Toxicity of Pesticides, Faculty of Agriculture, Kafrelsheikh University, Egypt.
| |
Collapse
|
26
|
Uridine treatment prevents REM sleep deprivation-induced learning and memory impairment. Neurosci Res 2019; 148:42-48. [PMID: 30685492 DOI: 10.1016/j.neures.2019.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/14/2018] [Accepted: 01/21/2019] [Indexed: 01/05/2023]
Abstract
Previous studies have shown that sleep plays an important role in cognitive functions and sleep deprivation impairs learning and memory. Uridine is the main pyrimidine nucleoside found in human blood circulation and has beneficial effects on cognitive functions. The aim of the present study was to investigate the effects of uridine administration on learning and memory impairment in sleep-deprived rats. Flower pot method was used to induce REM sleep deprivation. Uridine-treated groups received 1 mmol/kg uridine and control groups received 1 ml/kg saline (0.9% NaCl) twice a day for four days and once a day on the 5th day intraperitoneally. Learning and memory performances were measured using Morris water maze (MWM) test. We also measured the ratios of total calcium-calmodulin dependent kinase II (tCaMKII)/β-tubulin and phosphorylated cyclic adenosine monophosphate (cAMP) response element binding protein (pCREB)/β-tubulin, long-term potentiation (LTP) related molecules, using western blot analysis on the hippocampus. The results showed that REM sleep deprivation impaired learning and memory and also decreased the ratios of tCaMKII and pCREB. Uridine treatment enhanced learning and memory parameters in REM sleep-deprived rats. Additionally, decreases in tCaMKII and pCREB were prevented by uridine treatment. These data suggest that administration of uridine for five consecutive days prevents REM sleep deprivation-induced deficits in learning and memory associated with enhanced tCaMKII and pCREB ratios in the hippocampus.
Collapse
|
27
|
Wellman CL, Moench KM. Preclinical studies of stress, extinction, and prefrontal cortex: intriguing leads and pressing questions. Psychopharmacology (Berl) 2019; 236:59-72. [PMID: 30225660 PMCID: PMC6374178 DOI: 10.1007/s00213-018-5023-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 09/03/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Stress is associated with cognitive and emotional dysfunction, and increases risk for a variety of psychological disorders, including depression and posttraumatic stress disorder. Prefrontal cortex is critical for executive function and emotion regulation, is a target for stress hormones, and is implicated in many stress-influenced psychological disorders. Extinction of conditioned fear provides an excellent model system for examining how stress-induced changes in corticolimbic structure and function are related to stress-induced changes in neural function and behavior, as the neural circuitry underlying this behavior is well characterized. OBJECTIVES This review examines how acute and chronic stress influences extinction and describes how stress alters the structure and function of the medial prefrontal cortex, a potential neural substrate for these effects. In addition, we identify important unanswered questions about how stress-induced change in prefrontal cortex may mediate extinction deficits and avenues for future research. KEY FINDINGS A substantial body of work demonstrates deficits in extinction after either acute or chronic stress. A separate and substantial literature demonstrates stress-induced neuronal remodeling in medial prefrontal cortex, along with several key neurohormonal contributors to this remodeling, and there is substantial overlap in prefrontal mechanisms underlying extinction and the mechanisms implicated in stress-induced dysfunction of-and neuronal remodeling in-medial prefrontal cortex. However, data directly examining the contribution of changes in prefrontal structure and function to stress-induced extinction deficits is currently lacking. CONCLUSIONS Understanding how stress influences extinction and its neural substrates as well as individual differences in this effect will elucidate potential avenues for novel interventions for stress-sensitive disorders characterized by deficits in extinction.
Collapse
Affiliation(s)
- Cara L. Wellman
- Department of Psychological & Brain Sciences, Indiana University,Department of Psychological, Center for the Integrative Study of Animal Behavior, Indiana University,Department of Psychological, Program in Neuroscience, Indiana University
| | - Kelly M. Moench
- Department of Psychological & Brain Sciences, Indiana University,Department of Psychological, Center for the Integrative Study of Animal Behavior, Indiana University,Department of Psychological, Program in Neuroscience, Indiana University
| |
Collapse
|
28
|
Barfield ET, Gourley SL. Prefrontal cortical trkB, glucocorticoids, and their interactions in stress and developmental contexts. Neurosci Biobehav Rev 2018; 95:535-558. [PMID: 30477984 PMCID: PMC6392187 DOI: 10.1016/j.neubiorev.2018.10.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/14/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
The tropomyosin/tyrosine receptor kinase B (trkB) and glucocorticoid receptor (GR) regulate neuron structure and function and the hormonal stress response. Meanwhile, disruption of trkB and GR activity (e.g., by chronic stress) can perturb neuronal morphology in cortico-limbic regions implicated in stressor-related illnesses like depression. Further, several of the short- and long-term neurobehavioral consequences of stress depend on the developmental timing and context of stressor exposure. We review how the levels and activities of trkB and GR in the prefrontal cortex (PFC) change during development, interact, are modulated by stress, and are implicated in depression. We review evidence that trkB- and GR-mediated signaling events impact the density and morphology of dendritic spines, the primary sites of excitatory synapses in the brain, highlighting effects in adolescents when possible. Finally, we review the role of neurotrophin and glucocorticoid systems in stress-related metaplasticity. We argue that better understanding the long-term effects of developmental stressors on PFC trkB, GR, and related factors may yield insights into risk for chronic, remitting depression and related neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Elizabeth T Barfield
- Department of Pediatrics, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Graduate Program in Neuroscience, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.
| | - Shannon L Gourley
- Department of Pediatrics, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Graduate Program in Neuroscience, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Molecular and Systems Pharmacology Program, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.
| |
Collapse
|
29
|
Neuropathic Pain Creates an Enduring Prefrontal Cortex Dysfunction Corrected by the Type II Diabetic Drug Metformin But Not by Gabapentin. J Neurosci 2018; 38:7337-7350. [PMID: 30030404 DOI: 10.1523/jneurosci.0713-18.2018] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/10/2018] [Accepted: 07/13/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic pain patients suffer from pain-related cognitive deficits, even when taking commonly prescribed analgesics. These deficits are likely related to pain-related maladaptive plasticity in the frontal cortex. We sought to model cognitive deficits in mice with neuropathic pain to examine maladaptive morphological plasticity in the mPFC and to assess the effects of several therapeutics. We used an attentional set-shifting task in mice with spared nerve injury (SNI) who received either a single intrathecal injection of an analgesic dose of clonidine, 7 d of 100 mg/kg gabapentin, or 7 d of 200 mg/kg metformin. Male SNI mice were significantly more impaired in the set-shifting task than females. This deficit correlated with a loss of parvalbumin (PV) and reductions in axon initial segment (AIS) length in layers 5/6 of the infralimbic (IL) cortex. Acute pain relief with clonidine had no effect on set-shifting performance, whereas pain relief via 7 day treatment with gabapentin worsened the impairment in both SNI and sham mice. Gabapentin reversed the PV loss in the IL but had no effect on AIS length. Treatment with the AMPK-activator metformin completely reversed the pain-related cognitive impairment and restored AIS length in the IL but had little effect on PV expression. Our findings reveal that neuropathic pain-related cognitive impairments in male mice are correlated to bilateral morphological changes in PV interneurons and layer 5/6 IL pyramidal neuron AIS. Pain relief with metformin can reverse some of the functional and anatomical changes.SIGNIFICANCE STATEMENT Cognitive impairments are a comorbidity of neuropathic pain but are inadequately addressed by existing therapeutics. We used a neuropathic pain model in mice to demonstrate that male (but not female) mice show a robust pain-related deficit in attentional set-shifting, which is associated with structural plasticity in axon initial segments in the infralimbic cortex. These deficits were completely reversed by 7 day treatment with the antidiabetic drug metformin, suggesting that this drug can be repurposed for the treatment of neuropathic pain and its cognitive comorbidities. Our findings have implications for our understanding of how neuropathic pain causes structural plasticity in the brain, and they point to a marked sexual dimorphism in neuropathic pain mechanisms in mice.
Collapse
|
30
|
Homiack D, O'Cinneide E, Hajmurad S, Dohanich GP, Schrader LA. Effect of acute alarm odor exposure and biological sex on generalized avoidance and glutamatergic signaling in the hippocampus of Wistar rats. Stress 2018; 21:292-303. [PMID: 29916754 DOI: 10.1080/10253890.2018.1484099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is characterized by the development of paradoxical memory disturbances including intrusive memories and amnesia for specific details of the traumatic experience. Despite evidence that women are at higher risk to develop PTSD, most animal research has focused on the processes by which male rodents develop adaptive fear memory. As such, the mechanisms contributing to sex differences in the development of PTSD-like memory disturbances are poorly understood. In this investigation, we exposed adult male and female Wistar rats to the synthetic alarm odor 2,4,5-trimethylthiazole (TMT) to assess development of generalized fear behavior and rapid modulation of glutamate uptake and signaling cascades associated with hippocampus-dependent long-term memory. We report that female Wistar rats exposed to alarm odor exhibit context discrimination impairments relative to TMT-exposed male rats, suggesting the intriguing possibility that females are at greater risk in developing generalized fear memories. Mechanistically, alarm odor exposure rapidly modulated signaling cascades consistent with activation of the CREB shut-off cascade in the male, but not the female hippocampus. Moreover, TMT exposure dampened glutamate uptake and affected expression of the glutamate transporter, GLT-1 in the hippocampus. Taken together, these results provide evidence for rapid sex-dependent modulation of CREB signaling in the hippocampus by alarm odor exposure which may contribute to the development of generalized fear.
Collapse
Affiliation(s)
- Damek Homiack
- a Neuroscience Program, Brain Institute , Tulane University , New Orleans , LA , USA
| | - Emma O'Cinneide
- a Neuroscience Program, Brain Institute , Tulane University , New Orleans , LA , USA
| | - Sema Hajmurad
- b Department of Cell and Molecular Biology , Tulane University , New Orleans , LA , USA
| | - Gary P Dohanich
- a Neuroscience Program, Brain Institute , Tulane University , New Orleans , LA , USA
- c Department of Psychology , Tulane University , New Orleans , LA , USA
| | - Laura A Schrader
- a Neuroscience Program, Brain Institute , Tulane University , New Orleans , LA , USA
- b Department of Cell and Molecular Biology , Tulane University , New Orleans , LA , USA
| |
Collapse
|
31
|
Baratta MV, Leslie NR, Fallon IP, Dolzani SD, Chun LE, Tamalunas AM, Watkins LR, Maier SF. Behavioural and neural sequelae of stressor exposure are not modulated by controllability in females. Eur J Neurosci 2018; 47:959-967. [PMID: 29359831 PMCID: PMC5902414 DOI: 10.1111/ejn.13833] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/12/2022]
Abstract
The degree of behavioural control that an organism has over a stressor is a potent modulator of the stressor's impact; controllable stressors produce none of the neurochemical and behavioural sequelae that occur if the stressor is uncontrollable. Research demonstrating the importance of control and the neural mechanisms responsible has been conducted almost entirely with male rats. It is unknown if behavioural control is stress blunting in females, and whether or not a similar resilience circuitry is engaged. Female rats were exposed to controllable, yoked uncontrollable or no tailshock. In separate experiments, behavioural (juvenile social exploration, fear and shuttle box escape) and neurochemical (activation of dorsal raphe serotonin and dorsal raphe-projecting prelimbic neurons) outcomes, which are sensitive to the dimension of control in males, were assessed. Despite successful acquisition of the controlling response, behavioural control did not mitigate dorsal raphe serotonergic activation and behavioural outcomes induced by tailshock, as it does in males. Moreover, behavioural control failed to selectively engage prelimbic cells that project to the dorsal raphe as in males. Pharmacological activation of the prelimbic cortex restored the stress-buffering effects of control. Collectively, the data demonstrate stressor controllability phenomena are absent in females and that the protective prelimbic circuitry is present but not engaged. Reduced benefit from coping responses may represent a novel approach for understanding differential sex prevalence in stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Michael V. Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Nathan R. Leslie
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Isabella P. Fallon
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Samuel D. Dolzani
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado, USA
| | - Lauren E. Chun
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Andrew M. Tamalunas
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Linda R. Watkins
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Steven F. Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
32
|
Unpredictable chronic mild stress differentially impairs social and contextual discrimination learning in two inbred mouse strains. PLoS One 2017; 12:e0188537. [PMID: 29166674 PMCID: PMC5699833 DOI: 10.1371/journal.pone.0188537] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/08/2017] [Indexed: 12/28/2022] Open
Abstract
Alterations in the social and cognitive domain are considered important indicators for increased disability in many stress-related disorders. Similar impairments have been observed in rodents chronically exposed to stress, mimicking potential endophenotypes of stress-related psychopathologies such as major depression disorder (MDD), anxiety, conduct disorder, and posttraumatic stress disorder (PTSD). Data from numerous studies suggest that deficient plasticity mechanisms in hippocampus (HC) and prefrontal cortex (PFC) might underlie these social and cognitive deficits. Specifically, stress-induced deficiencies in neural plasticity have been associated with a hypodopaminergic state and reduced neural plasticity persistence. Here we assessed the effects of unpredictable chronic mild stress (UCMS) on exploratory, social and cognitive behavior of females of two inbred mouse strains (C57BL/6J and DBA/2J) that differ in their dopaminergic profile. Exposure to chronic stress resulted in impaired circadian rhythmicity, sociability and social cognition in both inbred strains, but differentially affected activity patterns and contextual discrimination performance. These stress-induced behavioral impairments were accompanied by reduced expression levels of brain derived neurotrophic factor (BDNF) in the prefrontal cortex. The strain-specific cognitive impairment was coexistent with enhanced plasma corticosterone levels and reduced expression of genes related to dopamine signaling in hippocampus. These results underline the importance of assessing different strains with multiple test batteries to elucidate the neural and genetic basis of social and cognitive impairments related to chronic stress.
Collapse
|
33
|
Homiack D, O'Cinneide E, Hajmurad S, Barrileaux B, Stanley M, Kreutz MR, Schrader LA. Predator odor evokes sex-independent stress responses in male and female Wistar rats and reduces phosphorylation of cyclic-adenosine monophosphate response element binding protein in the male, but not the female hippocampus. Hippocampus 2017; 27:1016-1029. [PMID: 28599071 DOI: 10.1002/hipo.22749] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/11/2017] [Accepted: 05/23/2017] [Indexed: 12/20/2022]
Abstract
Post-traumatic stress disorder (PTSD) is characterized by memory disturbances following trauma. Acute predator threat has emerged as an ethological model of PTSD, yet the effects of predator odor on signaling cascades associated with long-term memory remain poorly understood. In this study, we exposed male and female Wistar rats to the synthetic predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT) to assess behavioral and physiological responses as well as rapid modulation of signal transduction cascades associated with learning and memory in the male and female hippocampus. During exposure to TMT in the homecage, both male and female animals displayed robust immobility, avoidance, and altered activity as a function of time. Physiologically, TMT exposure increased circulating corticosterone and blood glucose in both male and female rodents, suggesting that TMT evokes sex-independent behavioral and physiological responses. With respect to signal transduction, TMT exposure rapidly reduced phosphorylation of cyclic-adenosine monophosphate response element binding protein (CREB) in the male, but not the female hippocampus. Furthermore, TMT exposure reduced phosphorylation of extracellular signal-regulated kinase 1/2 and increased nuclear expression of the synapto-nuclear messenger protein Jacob in the male hippocampus, consistent with activation of the CREB shut-off pathway. In a follow-up behavioral experiment, post-training exposure to TMT did not affect spatial water maze performance of male rats. However, male rats re-introduced to the context in which TMT had previously been presented displayed avoidance and hyperactivity, but not freezing behavior or elevated corticosterone responses, suggesting that TMT exposure supports a form of contextual conditioning which is not characterized by immobility. Taken together, our findings suggest that TMT evokes similar behavioral and physiological responses in male and female Wistar rats, but affects distinct signaling cascades in the male and female hippocampus which may contribute to behavioral disruptions associated with predator exposure.
Collapse
Affiliation(s)
- Damek Homiack
- Neuroscience Program, Brain Institute, Tulane University, New Orleans, Louisiana, 70118
| | - Emma O'Cinneide
- Neuroscience Program, Brain Institute, Tulane University, New Orleans, Louisiana, 70118
| | - Sema Hajmurad
- Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, 70118
| | - Brett Barrileaux
- Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, 70118
| | - Mary Stanley
- Neuroscience Program, Brain Institute, Tulane University, New Orleans, Louisiana, 70118.,Infectious Disease and Microbiome Program, Broad Institute, Cambridge, Massachusetts, 02142
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group 'Dendritic Organelles and Synaptic Function', Hamburg, Germany
| | - Laura A Schrader
- Neuroscience Program, Brain Institute, Tulane University, New Orleans, Louisiana, 70118.,Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, 70118
| |
Collapse
|
34
|
Saland SK, Duclot F, Kabbaj M. Integrative analysis of sex differences in the rapid antidepressant effects of ketamine in preclinical models for individualized clinical outcomes. Curr Opin Behav Sci 2016; 14:19-26. [PMID: 28584860 DOI: 10.1016/j.cobeha.2016.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In major depressive disorder, women exhibit higher lifetime prevalence and different antidepressant response rates than men, which illustrates the importance of examining individual differences in the pathophysiology of depression and therapeutic response. In recent years, the consideration of sex in related preclinical research has thus gained interest-particularly in light of novel evidence for rapid-acting antidepressants. Notably, the literature recently revealed a higher sensitivity of females to the antidepressant effects of the N-methyl-D-aspartate receptor antagonist ketamine, in both baseline and preclinical conditions. Combined with its fast-acting and relatively sustained properties, this evidence highlights ketamine as a particularly interesting therapeutic alternative for this sensitive population, and supports the value in considering sex as a critical factor for improved individualized therapeutic strategies.
Collapse
Affiliation(s)
- Samantha K Saland
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL-32306.,Program in Neuroscience, Florida State University, Tallahassee, FL-32306
| | - Florian Duclot
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL-32306.,Program in Neuroscience, Florida State University, Tallahassee, FL-32306
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL-32306.,Program in Neuroscience, Florida State University, Tallahassee, FL-32306
| |
Collapse
|
35
|
Sikandaner HE, Park SY, Kim MJ, Park SN, Yang DW. Neuroprotective effects of sildenafil against oxidative stress and memory dysfunction in mice exposed to noise stress. Behav Brain Res 2016; 319:37-47. [PMID: 27836585 DOI: 10.1016/j.bbr.2016.10.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/27/2016] [Accepted: 10/28/2016] [Indexed: 12/13/2022]
Abstract
Noise exposure has been well characterized as an environmental stressor, and is known to have auditory and non-auditory effects. Phosphodiesterase 5 (PDE5) inhibitors affect memory and hippocampus plasticity through various signaling cascades which are regulated by cGMP. In this study, we investigated the effects of sildenafil on memory deficiency, neuroprotection and oxidative stress in mice caused by chronic noise exposure. Mice were exposed to noise for 4h every day up to 14days at 110dB SPL of noise level. Sildenafil (15mg/kg) was orally administered 30min before noise exposure for 14days. Behavioral assessments were performed using novel object recognition (NOR) test and radial arm maze (RAM) test. Higher levels of memory dysfunction and oxidative stress were observed in noise alone-induced mice compared to control group. Interestingly, sildenafil administration increased memory performance, decreased oxidative stress, and increased neuroprotection in the hippocampus region of noise alone-induced mice likely through affecting memory related pathways such as cGMP/PKG/CREB and p25/CDK5, and induction of free radical scavengers such as SOD1, SOD2, SOD3, Prdx5, and catalase in the brain of stressed mice.
Collapse
Affiliation(s)
- Hu Erxidan Sikandaner
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - So Young Park
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Jung Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Shi Nae Park
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Dong Won Yang
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Gouweleeuw L, Hovens IB, Liu H, Naudé PJ, Schoemaker RG. Differences in the association between behavior and neutrophil gelatinase-associated lipocalin in male and female rats after coronary artery ligation. Physiol Behav 2016; 163:7-16. [DOI: 10.1016/j.physbeh.2016.04.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/05/2016] [Accepted: 04/22/2016] [Indexed: 01/06/2023]
|
37
|
Cossio R, Carreira MB, Vásquez CE, Britton GB. Sex differences and estrous cycle effects on foreground contextual fear conditioning. Physiol Behav 2016; 163:305-311. [PMID: 27195460 DOI: 10.1016/j.physbeh.2016.05.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 05/13/2016] [Accepted: 05/15/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Ricardo Cossio
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama
| | - María B Carreira
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama
| | - Carol E Vásquez
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama; Department of Biotechnology, AcharyaNagarjuna University, Guntur, India
| | - Gabrielle B Britton
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama.
| |
Collapse
|
38
|
Venezia AC, Guth LM, Sapp RM, Spangenburg EE, Roth SM. Sex-dependent and independent effects of long-term voluntary wheel running on Bdnf mRNA and protein expression. Physiol Behav 2016; 156:8-15. [PMID: 26752611 PMCID: PMC4753141 DOI: 10.1016/j.physbeh.2015.12.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/14/2015] [Accepted: 12/29/2015] [Indexed: 12/31/2022]
Abstract
UNLABELLED The beneficial effects of physical activity on brain health (synaptogenesis, neurogenesis, enhanced synaptic plasticity, improved learning and memory) appear to be mediated through changes in region-specific expression of neurotrophins, transcription factors, and postsynaptic receptors, though investigations of sex differences in response to long-term voluntary wheel running are limited. PURPOSE To examine the effect of five months of voluntary wheel running on hippocampal mRNA and protein expression of factors critical for exercise-induced structural and functional plasticity in male and female adult mice. METHODS At 8weeks of age, male and female C57BL/6 mice were individually housed with (PA; n=20; 10 male) or without (SED; n=20; 10 male) access to a computer monitored voluntary running wheel. At 28weeks, all mice were sacrificed and hippocampi removed. Total RNA was isolated from the hippocampus and expression of total Bdnf, Bdnf transcript IV, tPA, Pgc-1a, GluR1, NR2A, and NR2B were assessed with quantitative RT-PCR and total and mature Bdnf protein were assessed with ELISA. RESULTS We found significantly higher Bdnf IV mRNA expression in PA males (p=0.03) and females (p=0.03) compared to SED animals. Total Bdnf mRNA expression was significantly greater in PA males compared to SED males (p=0.01), but there was no difference in females. Similarly, we observed significantly higher mature Bdnf protein in PA males compared to SED males (p=0.04), but not in females. CONCLUSION These findings indicate that the impact of long-term voluntary wheel running on transcriptional and post-translational regulation of Bdnf may be sex-dependent, though the activity-dependent Bdnf IV transcript is sensitive to exercise independent of sex.
Collapse
Affiliation(s)
- Andrew C Venezia
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, USA; Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD, USA
| | - Lisa M Guth
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, USA
| | - Ryan M Sapp
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, USA
| | - Espen E Spangenburg
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, USA
| | - Stephen M Roth
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, USA; Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD, USA.
| |
Collapse
|
39
|
Joel D, Fausto-Sterling A. Beyond sex differences: new approaches for thinking about variation in brain structure and function. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150451. [PMID: 26833844 DOI: 10.1098/rstb.2015.0451] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2015] [Indexed: 12/21/2022] Open
Abstract
In the study of variation in brain structure and function that might relate to sex and gender, language matters because it frames our research questions and methods. In this article, we offer an approach to thinking about variation in brain structure and function that pulls us outside the sex differences formulation. We argue that the existence of differences between the brains of males and females does not unravel the relations between sex and the brain nor is it sufficient to characterize a population of brains. Such characterization is necessary for studying sex effects on the brain as well as for studying brain structure and function in general. Animal studies show that sex interacts with environmental, developmental and genetic factors to affect the brain. Studies of humans further suggest that human brains are better described as belonging to a single heterogeneous population rather than two distinct populations. We discuss the implications of these observations for studies of brain and behaviour in humans and in laboratory animals. We believe that studying sex effects in context and developing or adopting analytical methods that take into account the heterogeneity of the brain are crucial for the advancement of human health and well-being.
Collapse
Affiliation(s)
- Daphna Joel
- School of Psychological Sciences, Tel-Aviv University, Ramat Aviv, Tel-Aviv, Israel Sagol School of Neuoroscience, Tel-Aviv University, Ramat Aviv, Tel-Aviv, Israel
| | - Anne Fausto-Sterling
- Department of Molecular Biology, Cell and Biochemistry, Brown University, Providence, RI, USA
| |
Collapse
|
40
|
Sardari M, Rezayof A, Khodagholi F. Hippocampal signaling pathways are involved in stress-induced impairment of memory formation in rats. Brain Res 2015; 1625:54-63. [PMID: 26301822 DOI: 10.1016/j.brainres.2015.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/11/2015] [Accepted: 08/16/2015] [Indexed: 01/28/2023]
Abstract
Stress is a potent modulator of hippocampal-dependent memory formation. The aim of the present study was to assess the role of hippocampal signaling pathways in stress-induced memory impairment in male Wistar rats. The animals were exposed to acute elevated platform (EP) stress and memory formation was measured by a step-through type passive avoidance task. The results indicated that post-training or pre-test exposure to EP stress impaired memory consolidation or retrieval respectively. Using western blot analysis, it was found that memory retrieval was associated with the increase in the levels of phosphorylated cAMP-responsive element binding protein (P-CREB), peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and its downstream targets in the hippocampus. In contrast, the stress exposure decreased the hippocampal levels of these proteins. In addition, stress-induced impairment of memory consolidation or retrieval was associated with the decrease in the P-CREB/CREB ratio and the PGC-1α level in the hippocampus. On the other hand, the hippocampal level of nuclear factor E2-related factor 2 (Nrf2) and gamma-glutamylcysteine synthetase (γ-GCS) which are the master regulators of defense system were decreased by the stress exposure. The increased hippocampal levels of Nrf2 and it׳s downstream was observed during memory retrieval, while stress-induced impairment of memory consolidation or retrieval inhibited this hippocampal signaling pathway. Overall, these findings suggest that down-regulation of CREB/PGC-1α signaling cascade and Nrf2 antioxidant pathways in the hippocampus may be associated with memory impairment induced by stress.
Collapse
Affiliation(s)
- Maryam Sardari
- Department of Animal Biology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran.
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Differences in Active Avoidance Conditioning in Male and Female Rats with Experimental Anxiety-Depressive Disorder. Bull Exp Biol Med 2015. [PMID: 26201906 DOI: 10.1007/s10517-015-2956-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Using rat model of experimental anxiety-depressive disorder caused by postnatal administration of methionyl-2(S)-cyanopyrrolidine, an inhibitor of dipeptidyl peptidase IV, we compared conditioned active avoidance response and memory retention in males and females. In experimental males and females, conditioning was impaired in comparison with the control. In experimental groups, females were worse learners than males, while in control groups, females were better learners than males. Memory retention in experimental animals did not differ from that in controls 24 h after learning. Two months after learning, control females demonstrated better retention than control males.
Collapse
|
42
|
Moench KM, Wellman CL. Stress-induced alterations in prefrontal dendritic spines: Implications for post-traumatic stress disorder. Neurosci Lett 2014; 601:41-5. [PMID: 25529195 DOI: 10.1016/j.neulet.2014.12.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/09/2014] [Accepted: 12/16/2014] [Indexed: 10/24/2022]
Abstract
The medial prefrontal cortex (mPFC) is involved in a variety of important functions including emotional regulation, HPA axis regulation, and working memory. It also demonstrates remarkable plasticity in an experience-dependent manner. There is extensive evidence that stressful experiences can produce profound changes in the morphology of neurons within mPFC with a variety of behavioral consequences. The deleterious behavioral outcomes associated with mPFC dysfunction have been implicated in multiple psychopathologies, including post-traumatic stress disorder (PTSD). Given the prevalence of these disorders, a deeper understanding of the cellular mechanisms underlying stress-induced morphological changes in mPFC is critical, and could lead to improved therapeutic treatments. Here we give a brief review of recent studies examining the mechanisms underlying changes in mPFC pyramidal neuron dendritic spines - the primary sites of excitatory input in cortical pyramidal neurons. We begin with an overview of the effects of chronic stress on mPFC dendritic spine density and morphology followed by proposed mechanisms for these changes. We then discuss the time course of stress effects on mPFC as well as potential intercellular influences. Given that many psychopathologies, including PTSD, have different prevalence rates among men and women, we end with a discussion of the sex differences that have been observed in morphological changes in mPFC. Future directions and implications for PTSD are discussed throughout.
Collapse
Affiliation(s)
- Kelly M Moench
- Department of Psychological & Brain Sciences, Program in Neural Science, and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA
| | - Cara L Wellman
- Department of Psychological & Brain Sciences, Program in Neural Science, and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
43
|
Hudson S, Jacobson-Pick S, Anisman H. Sex differences in behavior and pro-inflammatory cytokine mRNA expression following stressor exposure and re-exposure. Neuroscience 2014; 277:239-49. [DOI: 10.1016/j.neuroscience.2014.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/16/2014] [Accepted: 07/07/2014] [Indexed: 12/22/2022]
|
44
|
Shin S, Le Lay J, Everett LJ, Gupta R, Rafiq K, Kaestner KH. CREB mediates the insulinotropic and anti-apoptotic effects of GLP-1 signaling in adult mouse β-cells. Mol Metab 2014; 3:803-12. [PMID: 25379405 PMCID: PMC4216406 DOI: 10.1016/j.molmet.2014.08.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 08/12/2014] [Accepted: 08/15/2014] [Indexed: 12/27/2022] Open
Abstract
Objective Glucagon-like peptide-1 (GLP-1) plays a major role in pancreatic β-cell function and survival by increasing cytoplasmic cAMP levels, which are thought to affect transcription through activation of the basic leucine zipper (bZIP) transcription factor CREB. Here, we test CREB function in the adult β-cell through inducible gene deletion. Methods We employed cell type-specific and inducible gene ablation to determine CREB function in pancreatic β-cells in mice. Results By ablating CREB acutely in mature β-cells in tamoxifen-treated CrebloxP/loxP;Pdx1-CreERT2 mice, we show that CREB has little impact on β-cell turnover, in contrast to what had been postulated previously. Rather, CREB is required for GLP-1 to elicit its full effects on stimulating glucose-induced insulin secretion and protection from cytokine-induced apoptosis. Mechanistically, we find that CREB regulates expression of the pro-apoptotic gene p21 (Cdkn1a) in β-cells, thus demonstrating that CREB is essential to mediating this critical aspect of GLP-1 receptor signaling. Conclusions In sum, our studies using conditional gene deletion put into question current notions about the importance of CREB in regulating β-cell function and mass. However, we reveal an important role for CREB in the β-cell response to GLP-1 receptor signaling, further validating CREB as a therapeutic target for diabetes.
Collapse
Affiliation(s)
- Soona Shin
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - John Le Lay
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Logan J Everett
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Rana Gupta
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Kiran Rafiq
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
45
|
Joel D, Yankelevitch-Yahav R. Reconceptualizing sex, brain and psychopathology: interaction, interaction, interaction. Br J Pharmacol 2014; 171:4620-35. [PMID: 24758640 DOI: 10.1111/bph.12732] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 03/22/2014] [Accepted: 03/26/2014] [Indexed: 01/08/2023] Open
Abstract
In recent years there has been a growing recognition of the influence of sex on brain structure and function, and in relation, on the susceptibility, prevalence and response to treatment of psychiatric disorders. Most theories and descriptions of the effects of sex on the brain are dominated by an analogy to the current interpretation of the effects of sex on the reproductive system, according to which sex is a divergence system that exerts a unitary, overriding and serial effect on the form of other systems. We shortly summarize different lines of evidence that contradict aspects of this analogy. The new view that emerges from these data is of sex as a complex system whose different components interact with one another and with other systems to affect body and brain. The paradigm shift that this understanding calls for is from thinking of sex in terms of sexual dimorphism and sex differences, to thinking of sex in terms of its interactions with other factors and processes. Our review of data obtained from animal models of psychopathology clearly reveals the need for such a paradigmatic shift, because in the field of animal behaviour whether a sex difference exists and its direction depend on the interaction of many factors including, species, strain, age, specific test employed and a multitude of environmental factors. We conclude by explaining how the new conceptualization can account for sex differences in psychopathology.
Collapse
Affiliation(s)
- D Joel
- School of Psychological Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
46
|
Clark SM, Sand J, Francis TC, Nagaraju A, Michael KC, Keegan AD, Kusnecov A, Gould TD, Tonelli LH. Immune status influences fear and anxiety responses in mice after acute stress exposure. Brain Behav Immun 2014; 38:192-201. [PMID: 24524915 PMCID: PMC3989422 DOI: 10.1016/j.bbi.2014.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/27/2014] [Accepted: 02/01/2014] [Indexed: 12/15/2022] Open
Abstract
Significant evidence suggests that exposure to traumatic and/or acute stress in both mice and humans results in compromised immune function that in turn may affect associated brain processes. Additionally, recent studies in mouse models of immune deficiency have suggested that adaptive immunity may play a role during traumatic stress exposure and that impairments in lymphocyte function may contribute to increased susceptibility to various psychogenic stressors. However, rodent studies on the relationship between maladaptive stress responses and lymphocyte deficiency have been complicated by the fact that genetic manipulations in these models may also result in changes in CNS function due to the expression of targeted genes in tissues other than lymphocytes, including the brain. To address these issues we utilized mice with a deletion of recombination-activating gene 2 (Rag2), which has no confirmed expression in the CNS; thus, its loss should result in the absence of mature lymphocytes without altering CNS function directly. Stress responsiveness of immune deficient Rag2(-/-) mice on a BALB/c background was evaluated in three different paradigms: predator odor exposure (POE), fear conditioning (FC) and learned helplessness (LH). These models are often used to study different aspects of stress responsiveness after the exposure to an acute stressor. In addition, immunoblot analysis was used to assess hippocampal BDNF expression under both stressed and non-stressed conditions. Subsequent to POE, Rag2(-/-) mice exhibited a reduced acoustic startle response compared to BALB/c mice; no significant differences in behavior were observed in either FC or LH. Furthermore, analysis of hippocampal BDNF indicated that Rag2(-/-) mice have elevated levels of the mature form of BDNF compared to BALB/c mice. Results from our studies suggest that the absence of mature lymphocytes is associated with increased resilience to stress exposure in the POE and does not affect behavioral responses in the FC and LH paradigms. These findings indicate that lymphocytes play a specific role in stress responsiveness dependent upon the type, nature and intensity of the stressor.
Collapse
Affiliation(s)
- Sarah M Clark
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, United States
| | - Joseph Sand
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - T Chase Francis
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Anitha Nagaraju
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kerry C Michael
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Achsah D Keegan
- Center for Vascular and Inflammatory Diseases, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, United States
| | | | - Todd D Gould
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States; Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, United States; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, United States
| | - Leonardo H Tonelli
- Laboratory of Behavioral Neuroimmunology, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States; Research and Development Service, Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, United States.
| |
Collapse
|
47
|
Lucassen PJ, Pruessner J, Sousa N, Almeida OFX, Van Dam AM, Rajkowska G, Swaab DF, Czéh B. Neuropathology of stress. Acta Neuropathol 2014; 127:109-35. [PMID: 24318124 PMCID: PMC3889685 DOI: 10.1007/s00401-013-1223-5] [Citation(s) in RCA: 290] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 11/26/2013] [Indexed: 02/06/2023]
Abstract
Environmental challenges are part of daily life for any individual. In fact, stress appears to be increasingly present in our modern, and demanding, industrialized society. Virtually every aspect of our body and brain can be influenced by stress and although its effects are partly mediated by powerful corticosteroid hormones that target the nervous system, relatively little is known about when, and how, the effects of stress shift from being beneficial and protective to becoming deleterious. Decades of stress research have provided valuable insights into whether stress can directly induce dysfunction and/or pathological alterations, which elements of stress exposure are responsible, and which structural substrates are involved. Using a broad definition of pathology, we here review the "neuropathology of stress" and focus on structural consequences of stress exposure for different regions of the rodent, primate and human brain. We discuss cytoarchitectural, neuropathological and structural plasticity measures as well as more recent neuroimaging techniques that allow direct monitoring of the spatiotemporal effects of stress and the role of different CNS structures in the regulation of the hypothalamic-pituitary-adrenal axis in human brain. We focus on the hypothalamus, hippocampus, amygdala, nucleus accumbens, prefrontal and orbitofrontal cortex, key brain regions that not only modulate emotions and cognition but also the response to stress itself, and discuss disorders like depression, post-traumatic stress disorder, Cushing syndrome and dementia.
Collapse
Affiliation(s)
- Paul J. Lucassen
- SILS-Center for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Jens Pruessner
- Department of Psychiatry, Douglas Institute, McGill University, Montreal, QC Canada
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | | | - Anne Marie Van Dam
- Department of Anatomy and Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | - Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS USA
| | - Dick F. Swaab
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Boldizsár Czéh
- Department of Laboratory Medicine, Faculty of Medicine, University of Pécs, Pécs, Hungary
- Szentágothai János Research Center, Neuroendocrinology Research Group, University of Pécs, Pécs, Hungary
| |
Collapse
|
48
|
Lin M, Fwu PT, Buss C, Davis EP, Head K, Muftuler LT, Sandman CA, Su MY. Developmental changes in hippocampal shape among preadolescent children. Int J Dev Neurosci 2013; 31:473-81. [PMID: 23773912 DOI: 10.1016/j.ijdevneu.2013.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 06/03/2013] [Accepted: 06/04/2013] [Indexed: 11/25/2022] Open
Abstract
It is known that the largest developmental changes in the hippocampus take place during the prenatal period and during the first two years of postnatal life. Few studies have been conducted to address the normal developmental trajectory of the hippocampus during childhood. In this study shape analysis was applied to study the normal developing hippocampus in a group of 103 typically developing 6- to 10-year-old preadolescent children. The individual brain was normalized to a template, and then the hippocampus was manually segmented and further divided into the head, body, and tail sub-regions. Three different methods were applied for hippocampal shape analysis: radial distance mapping, surface-based template registration using the robust point matching (RPM) algorithm, and volume-based template registration using the Demons algorithm. All three methods show that the older children have bilateral expanded head segments compared to the younger children. The results analyzed based on radial distance to the centerline were consistent with those analyzed using template-based registration methods. In analyses stratified by sex, it was found that the age-associated anatomical changes were similar in boys and girls, but the age-association was strongest in girls. Total hippocampal volume and sub-regional volumes analyzed using manual segmentation did not show a significant age-association. Our results suggest that shape analysis is sensitive to detect sub-regional differences that are not revealed in volumetric analysis. The three methods presented in this study may be applied in future studies to investigate the normal developmental trajectory of the hippocampus in children. They may be further applied to detect early deviations from the normal developmental trajectory in young children for evaluating susceptibility for psychopathological disorders involving hippocampus.
Collapse
Affiliation(s)
- Muqing Lin
- Tu & Yuen Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, Irvine, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Marsden WN. Synaptic plasticity in depression: molecular, cellular and functional correlates. Prog Neuropsychopharmacol Biol Psychiatry 2013; 43:168-84. [PMID: 23268191 DOI: 10.1016/j.pnpbp.2012.12.012] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 12/14/2012] [Accepted: 12/15/2012] [Indexed: 12/31/2022]
Abstract
Synaptic plasticity confers environmental adaptability through modification of the connectivity between neurons and neuronal circuits. This is achieved through changes to synapse-associated signaling systems and supported by complementary changes to cellular morphology and metabolism within the tripartite synapse. Mounting evidence suggests region-specific changes to synaptic form and function occur as a result of chronic stress and in depression. Within subregions of the prefrontal cortex (PFC) and hippocampus structural and synapse-related findings seem consistent with a deficit in long-term potentiation (LTP) and facilitation of long-term depression (LTD), particularly at excitatory pyramidal synapses. Other brain regions are less well-studied; however the amygdala may feature a somewhat opposite synaptic pathology including reduced inhibitory tone. Changes to synaptic plasticity in stress and depression may correlate those to several signal transduction pathways (e.g. NOS-NO, cAMP-PKA, Ras-ERK, PI3K-Akt, GSK-3, mTOR and CREB) and upstream receptors (e.g. NMDAR, TrkB and p75NTR). Deficits in synaptic plasticity may further correlate disrupted brain redox and bioenergetics. Finally, at a functional level region-specific changes to synaptic plasticity in depression may relate to maladapted neurocircuitry and parallel reduced cognitive control over negative emotion.
Collapse
Affiliation(s)
- W N Marsden
- Highclere Court, Woking, Surrey, GU21 2QP, UK.
| |
Collapse
|
50
|
Bath KG, Schilit A, Lee FS. Stress effects on BDNF expression: Effects of age, sex, and form of stress. Neuroscience 2013; 239:149-56. [PMID: 23402850 DOI: 10.1016/j.neuroscience.2013.01.074] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/17/2012] [Accepted: 01/31/2013] [Indexed: 12/14/2022]
Affiliation(s)
- K G Bath
- Department of Neuroscience, Brown University, Box GL-N, 185 Meeting Street, Providence, RI 02912, USA.
| | | | | |
Collapse
|