1
|
Isgate SB, Budge KE, Byrnes EM, Vassoler FM. Paternal Morphine Alters Offspring Circulating Beta-Endorphin and Corticosterone Responses to Oxycodone and Cocaine. Neuropharmacology 2024; 265:110271. [PMID: 39694232 DOI: 10.1016/j.neuropharm.2024.110271] [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: 10/04/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND The opioid epidemic is leading to increased opioid use in adolescent populations. A growing body of evidence suggests that taking opioids during adolescence can disrupt normal development and impact future offspring. This study investigates the impact of paternal morphine exposure during adolescence on the hypothalamic-pituitary-adrenal (HPA) axis and release of endorphins in the offspring. METHODS Male rats were administered morphine once a day from postnatal day (PND)30-39 using an increasing dosing regimen (5-25mg/kg/day increasing every other day). They were mated during adulthood to drug naïve females. Their offspring were assessed for circulating beta-endorphin (βE) and corticosterone levels on PND30 (a timepoint prior to puberty in both sexes) in response to an acute injection of saline, oxycodone (1 mg/kg, i.p.) or cocaine (10 mg/kg, i.p.). At PND60, naïve littermates were catheterized so that a within-subjects design could be implemented to measure βE and corticosterone in response to saline, oxycodone, or cocaine. RESULTS In males, βE levels in the plasma were increased in Mor-F1 males compared to Sal-F1 males regardless of the acute injection. This elevation was observed at PND30 and PND60. There were no differences in female circulating βE. In terms of corticosterone, male Mor-F1 offspring had blunted corticosterone at PND30, but elevated corticosterone in response to oxycodone at PND60. The females also tended towards lower corticosterone prior to puberty but had significantly elevated levels of circulating corticosterone following an acute cocaine injection. CONCLUSION Paternal morphine exposure during adolescence induces sex- and drug-specific changes in secreted hormone responses in offspring. The alterations in βE and corticosterone levels suggest mechanisms through which adolescent opioid exposure can impact endocrine functions of future offspring. These findings contribute to the understanding of intergenerational transmission of substance use effects.
Collapse
Affiliation(s)
- Sara B Isgate
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - Kerri E Budge
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - Elizabeth M Byrnes
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - Fair M Vassoler
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA.
| |
Collapse
|
2
|
Shaw GA, Wegener AJ, Neigh GN. Chronic corticosterone administration alters synaptic mitochondrial function within the hippocampus of C57Bl/6NTac mice. Physiol Behav 2024; 287:114681. [PMID: 39209050 DOI: 10.1016/j.physbeh.2024.114681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/02/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Chronic activation of the hypothalamic-pituitary-adrenal axis increases circulating corticosterone levels, causing a host of downstream behavioral, molecular, and metabolic changes. Here, we assess the effects of chronic exogenous CORT administration on changes in behavior and mitochondrial respiration in hippocampal synaptosomes of male and female mice. Adult male (n = 15) and female (n = 17) C57Bl/6NTac mice were given 35ug/mL CORT or vehicle dissolved in their drinking water for 21 consecutive days. Chronic CORT increased piloerection in males only. Although volume of CORT-containing water consumed was similar between males and females, circulating plasma and fecal corticosterone levels were only elevated in CORT-exposed males. Behavioral effects of CORT were evident in the Y-maze such that CORT caused a decrease in direct revisits in both sexes. There was no observed presentation of anxiety-like behavior following chronic CORT administration. Functional hippocampal synaptosomes were analyzed for mitochondrial respiration using Agilent's Cell Mito Stress test. Chronic CORT caused a decrease in synaptic mitochondria basal respiration, maximal respiration, proton leak, and ATP production in both sexes. Despite only observing an effect of chronic CORT on corticosterone concentrations in fecal and blood samples of males, chronic CORT induced marked changes in hippocampal synaptic mitochondrial function of both sexes. These data highlight the importance of considering effects of stress hormone exposure on neural function even in the absence of measurable peripheral elevations in females.
Collapse
Affiliation(s)
- Gladys A Shaw
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy J Wegener
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Gretchen N Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
3
|
Fiamingo M, Bailey A, Toler S, Lee K, Oshiro W, Yoo B, Krantz T, Evansky P, Davies D, Gilmour MI, Farraj A, Jaspers I, Hazari MS. Enriched housing differentially alters allostatic load and cardiopulmonary responses to wildfire-related smoke in male and female mice. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:561-578. [PMID: 38721998 PMCID: PMC11167957 DOI: 10.1080/15287394.2024.2346582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Living conditions are an important modifier of individual health outcomes and may lead to higher allostatic load (AL). However, housing-induced cardiovascular and immune effects contributing to altered environmental responsiveness remain understudied. This investigation was conducted to examine the influence of enriched (EH) versus depleted housing (DH) conditions on cardiopulmonary functions, systemic immune responses, and allostatic load in response to a single wildfire smoke (WS) exposure in mice. Male and female C57BL/6J mice were divided into EH or DH for 22 weeks, and cardiopulmonary assessments measured before and after exposures to either one-hr filtered air (FA) or flaming eucalyptus WS exposure. Male and female DH mice exhibited increased heart rate (HR) and left ventricular mass (LVM), as well as reduced stroke volume and end diastolic volume (EDV) one week following exposure to WS. Female DH mice displayed significantly elevated levels of IL-2, IL-17, corticosterone and hemoglobin A1c (HbA1c) following WS, while female in EH mice higher epinephrine levels were detected. Female mice exhibited higher AL than males with DH, which was potentiated post-WS exposure. Thus, DH increased susceptibility to extreme air pollution in a gender-dependent manner suggesting that living conditions need to be evaluated as a modifier of toxicological responses.
Collapse
Affiliation(s)
- Michelle Fiamingo
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina – Chapel Hill, Chapel Hill, NC 27599
| | - Aleah Bailey
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina – Chapel Hill, Chapel Hill, NC 27599
| | - Sydnie Toler
- Gillings School of Global Public Health and School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaleb Lee
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830
| | - Wendy Oshiro
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Brendan Yoo
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Todd Krantz
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Paul Evansky
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - David Davies
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - M. Ian Gilmour
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Aimen Farraj
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Ilona Jaspers
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina – Chapel Hill, Chapel Hill, NC 27599
| | - Mehdi S. Hazari
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| |
Collapse
|
4
|
Park SB, Lur G. Repeated exposure to multiple concurrent stressors alters visual processing in the adult posterior parietal cortex. Neurobiol Stress 2024; 31:100660. [PMID: 39100726 PMCID: PMC11296072 DOI: 10.1016/j.ynstr.2024.100660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/25/2024] [Accepted: 06/29/2024] [Indexed: 08/06/2024] Open
Abstract
Chronic stress is well known to erode cognitive functions. Yet, our understanding of how repeated stress exposure impacts one of the fundamental bases of cognition: sensory processing, remains limited. The posterior parietal cortex (PPC) is a high order visual region, known for its role in visually guided decision making, multimodal integration, attention, and working memory. Here, we used functional measures to determine how repeated exposure to multiple concurrent stressors (RMS) affects sensory processing in the PPC in adult male mice. A longitudinal experimental design, repeatedly surveying the same population of neurons using in vivo two-photon imaging, revealed that RMS disrupts the balanced turnover of visually responsive cells in layer 2/3 of the PPC. Across the population, RMS-induced changes in visual responsiveness followed a bimodal distribution suggesting idiosyncratic stress effects. In cells that maintained their responsiveness across recording sessions, we found that stress reduced visual response magnitudes and feature selectivity. While we did not observe stress-induced elimination of excitatory synapses, noise correlation statistics indicated that RMS altered visual input to the neuronal population. The impact of RMS was restricted to visually evoked responses and was not evident in neuronal activity associated with locomotion onset. Together, our results indicate that despite no apparent synaptic reorganization, stress exposure in adulthood can disrupt sensory processing in the PPC, with the effects showing remarkable individual variation.
Collapse
Affiliation(s)
- Soo Bin Park
- Department of Neurobiology and Behavior, University of California, Irvine, CA USA, 92697
| | - Gyorgy Lur
- Department of Neurobiology and Behavior, University of California, Irvine, CA USA, 92697
| |
Collapse
|
5
|
Iqbal J, Huang GD, Xue YX, Yang M, Jia XJ. Role of estrogen in sex differences in memory, emotion and neuropsychiatric disorders. Mol Biol Rep 2024; 51:415. [PMID: 38472517 DOI: 10.1007/s11033-024-09374-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/22/2024] [Indexed: 03/14/2024]
Abstract
Estrogen regulates a wide range of neuronal functions in the brain, such as dendritic spine formation, remodeling of synaptic plasticity, cognition, neurotransmission, and neurodevelopment. Estrogen interacts with intracellular estrogen receptors (ERs) and membrane-bound ERs to produce its effect via genomic and non-genomic pathways. Any alterations in these pathways affect the number, size, and shape of dendritic spines in neurons associated with psychiatric diseases. Increasing evidence suggests that estrogen fluctuation causes changes in dendritic spine density, morphology, and synapse numbers of excitatory and inhibitory neurons differently in males and females. In this review, we discuss the role of estrogen hormone in rodents and humans based on sex differences. First, we explain estrogen role in learning and memory and show that a high estrogen level alleviates the deficits in learning and memory. Secondly, we point out that estrogen produces a striking difference in emotional memories in men and women, which leads them to display sex-specific differences in underlying neuronal signaling. Lastly, we discuss that fluctuations in estrogen levels in men and women are related to neuropsychiatric disorders, including schizophrenia, autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), bipolar disorder (BPD), major depressive disorder (MDD), substance use disorder (SUD), and anxiety disorders.
Collapse
Affiliation(s)
- Javed Iqbal
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No. 77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China
| | - Geng-Di Huang
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No. 77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Mei Yang
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No. 77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China.
- Clinical College of Mental Health, Shenzhen University Health Science Center, Shenzhen, China.
- Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, China.
| | - Xiao-Jian Jia
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, No. 77 Zhenbi Road, Pingshan District, Shenzhen, 518118, Guangdong, China.
- Clinical College of Mental Health, Shenzhen University Health Science Center, Shenzhen, China.
- Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
6
|
Gore IR, Gould E. Developmental and adult stress: effects of steroids and neurosteroids. Stress 2024; 27:2317856. [PMID: 38563163 PMCID: PMC11046567 DOI: 10.1080/10253890.2024.2317856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/03/2024] [Indexed: 04/04/2024] Open
Abstract
In humans, exposure to early life adversity has profound implications for susceptibility to developing neuropsychiatric disorders later in life. Studies in rodents have shown that stress experienced during early postnatal life can have lasting effects on brain development. Glucocorticoids and sex steroids are produced in endocrine glands and the brain from cholesterol; these molecules bind to nuclear and membrane-associated steroid receptors. Unlike other steroids that can also be made in the brain, neurosteroids bind specifically to neurotransmitter receptors, not steroid receptors. The relationships among steroids, neurosteroids, and stress are multifaceted and not yet fully understood. However, studies demonstrating altered levels of progestogens, androgens, estrogens, glucocorticoids, and their neuroactive metabolites in both developmental and adult stress paradigms strongly suggest that these molecules may be important players in stress effects on brain circuits and behavior. In this review, we discuss the influence of developmental and adult stress on various components of the brain, including neurons, glia, and perineuronal nets, with a focus on sex steroids and neurosteroids. Gaining an enhanced understanding of how early adversity impacts the intricate systems of brain steroid and neurosteroid regulation could prove instrumental in identifying novel therapeutic targets for stress-related conditions.
Collapse
Affiliation(s)
- Isha R Gore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| |
Collapse
|
7
|
Wang J, Akbari A, Chardahcherik M, Wu J. Ginger (Zingiber Officinale Roscoe) ameliorates ethanol-induced cognitive impairment by modulating NMDA and GABA-A receptors in rat hippocampus. Metab Brain Dis 2024; 39:67-76. [PMID: 37966694 DOI: 10.1007/s11011-023-01301-8] [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: 04/13/2023] [Accepted: 09/24/2023] [Indexed: 11/16/2023]
Abstract
Brain damage caused by ethanol abuse may lead to permanent damage, including severe dementia. The aim of this study was to investigate the effects of ginger powder on ethanol-induced cognitive disorders by examining oxidative damage and inflammation status, and the gene expression of N-methyl-D-aspartate (NMDA) and γ-Aminobutyric acid (GABA)-A receptors in the hippocampus of male rats. 24 adult male Sprague-Dawley rats were allocated randomly to four groups as follows control, ethanol (4g/kg/day, by gavage), ginger (1g/kg/day, by gavage), and ginger-ethanol. At the end of the study, memory and learning were evaluated by the shuttle box test. Moreover, to explore mechanisms involved in ethanol-induced cognitive impairment and the protective effect of ginger, the expression of Nuclear factor kappa B (NF-κB), nuclear factor erythroid 2-related factor 2 (Nrf2), NMDA receptor, and GABA-A receptor was measured along with inflammatory and oxidative biomarkers in the hippocampus tissue. The results showed that ethanol could induce cognitive impairment in the ethanol group, while pretreatment with ginger could reverse it. The gene expression of the NF-κB/ Tumor necrosis factor (TNF)-α/Interleukin (IL)-1β pathway and NMDA and GABA-A receptors significantly increased in the ethanol group compared to the control group. While pretreatment with ginger could significantly improve ethanol-induced cognitive impairment through these pathways in the ginger-ethanol group compared to the ethanol group (P < 0.05). It can be concluded that ginger powder could ameliorate ethanol-induced cognitive impairment by modulating the expression of NMDA and GABA-A receptors and inhibiting oxidative damage and the NF-κB/TNF-α/IL-1β pathway in the rat hippocampus.
Collapse
Affiliation(s)
- Jiaojiao Wang
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, 750000, China
| | - Abolfazl Akbari
- Department of Physiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Marjan Chardahcherik
- Department of Biochemistry, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Jun Wu
- Department of Internal Medicine, Xi'an Yanta Qiangsen Meilin Hospital, Xi'an, 710000, China.
| |
Collapse
|
8
|
Palamarchuk IS, Slavich GM, Vaillancourt T, Rajji TK. Stress-related cellular pathophysiology as a crosstalk risk factor for neurocognitive and psychiatric disorders. BMC Neurosci 2023; 24:65. [PMID: 38087196 PMCID: PMC10714507 DOI: 10.1186/s12868-023-00831-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
In this narrative review, we examine biological processes linking psychological stress and cognition, with a focus on how psychological stress can activate multiple neurobiological mechanisms that drive cognitive decline and behavioral change. First, we describe the general neurobiology of the stress response to define neurocognitive stress reactivity. Second, we review aspects of epigenetic regulation, synaptic transmission, sex hormones, photoperiodic plasticity, and psychoneuroimmunological processes that can contribute to cognitive decline and neuropsychiatric conditions. Third, we explain mechanistic processes linking the stress response and neuropathology. Fourth, we discuss molecular nuances such as an interplay between kinases and proteins, as well as differential role of sex hormones, that can increase vulnerability to cognitive and emotional dysregulation following stress. Finally, we explicate several testable hypotheses for stress, neurocognitive, and neuropsychiatric research. Together, this work highlights how stress processes alter neurophysiology on multiple levels to increase individuals' risk for neurocognitive and psychiatric disorders, and points toward novel therapeutic targets for mitigating these effects. The resulting models can thus advance dementia and mental health research, and translational neuroscience, with an eye toward clinical application in cognitive and behavioral neurology, and psychiatry.
Collapse
Affiliation(s)
- Iryna S Palamarchuk
- Centre for Addiction and Mental Health, 1001 Queen Street West, Toronto, ON, M6J1H4, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Sunnybrook Health Sciences Centre, Division of Neurology, Toronto, ON, Canada.
- Temerty Faculty of Medicine, Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada.
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tracy Vaillancourt
- Counselling Psychology, Faculty of Education, University of Ottawa, Ottawa, ON, Canada
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Tarek K Rajji
- Centre for Addiction and Mental Health, 1001 Queen Street West, Toronto, ON, M6J1H4, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Lipatova O, Campolattaro MM, Lockhart BK, Hammad MB. Differential effects of acute stress on spatial learning and memory in the open-field tower maze across the female estrous cycle. Neurobiol Learn Mem 2023; 206:107862. [PMID: 37944635 DOI: 10.1016/j.nlm.2023.107862] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
The purpose of the present investigation was to test how acute stress and levels of circulating estrogens together influence acquisition and retention of spatial learning, as well as explorative behaviors in female rats. We used the hippocampus-dependent Open-field Tower Maze (OFTM) task to assess acquisition followed by a retention test (reacquisition) that was given 48 h later. Immediately prior to acquisition, experimental rats were exposed to an acute restraint stress and were trained under bright lights. Female rats' estrous cycles were tracked throughout training and testing. Exposure to stress did not affect learning when levels of estrogens were low (i.e., during estrus and metestrus). However, acute stress exposure significantly lowered spatial acquisition of the female rats in the phases with rising levels of estrogens (i.e., during diestrus and proestrus). Furthermore, this stress-induced diminishment during acquisition was evident at the beginning of the retention without any presentation of stress. The present findings provide insight about the interactive relationship between stress and sex hormones on cognitive functions.
Collapse
Affiliation(s)
- Olga Lipatova
- Christopher Newport University, Newport News, VA, United States.
| | | | - Blakely K Lockhart
- Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Mariam B Hammad
- Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
10
|
Bjornson KJ, Vanderplow AM, Yang Y, Anderson DR, Kermath BA, Cahill ME. Stress-mediated dysregulation of the Rap1 small GTPase impairs hippocampal structure and function. iScience 2023; 26:107566. [PMID: 37664580 PMCID: PMC10470260 DOI: 10.1016/j.isci.2023.107566] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/15/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
The effects of repeated stress on cognitive impairment are thought to be mediated, at least in part, by reductions in the stability of dendritic spines in brain regions critical for proper learning and memory, including the hippocampus. Small GTPases are particularly potent regulators of dendritic spine formation, stability, and morphology in hippocampal neurons. Through the use of small GTPase protein profiling in mice, we identify increased levels of synaptic Rap1 in the hippocampal CA3 region in response to escalating, intermittent stress. We then demonstrate that increased Rap1 in the CA3 is sufficient in and of itself to produce stress-relevant dendritic spine and cognitive phenotypes. Further, using super-resolution imaging, we investigate how the pattern of Rap1 trafficking to synapses likely underlies its effects on the stability of select dendritic spine subtypes. These findings illuminate the involvement of aberrant Rap1 regulation in the hippocampus in contributing to the psychobiological effects of stress.
Collapse
Affiliation(s)
- Kathryn J. Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amanda M. Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yezi Yang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Danielle R. Anderson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Bailey A. Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael E. Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
11
|
Smiley CE, Pate BS, Bouknight SJ, Francis MJ, Nowicki AV, Harrington EN, Wood SK. Estrogen receptor beta in the central amygdala regulates the deleterious behavioral and neuronal consequences of repeated social stress in female rats. Neurobiol Stress 2023; 23:100531. [PMID: 36879670 PMCID: PMC9984877 DOI: 10.1016/j.ynstr.2023.100531] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/02/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
While over 95% of the population has reported experiencing extreme stress or trauma, females of reproductive age develop stress-induced neuropsychiatric disorders at twice the rate of males. This suggests that ovarian hormones may facilitate neural processes that increase stress susceptibility and underlie the heightened rates of these disorders, like depression and anxiety, that result from stress exposure in females. However, there is contradicting evidence in the literature regarding estrogen's role in stress-related behavioral outcomes. Estrogen signaling through estrogen receptor beta (ERβ) has been traditionally thought of as anxiolytic, but recent studies suggest estrogen exhibits distinct effects in the context of stress. Furthermore, ERβ is found abundantly in many stress-sensitive brain loci, including the central amygdala (CeA), in which transcription of the vital stress hormone, corticotropin releasing factor (CRF), can be regulated by an estrogen response element. Therefore, these experiments sought to identify the role of CeA ERβ activity during stress on behavioral outcomes in naturally cycling, adult, female Sprague-Dawley rats. Rats were exposed to an ethological model of vicarious social stress, witness stress (WS), in which they experienced the sensory and psychological aspects of an aggressive social defeat encounter between two males. Following WS, rats exhibited stress-induced anxiety-like behaviors in the marble burying taskand brain analysis revealed increased ERβ and CRF specifically within the CeA following exposure to stress cues. Subsequent experiments were designed to target this receptor in the CeA using microinjections of the ERβ antagonist, PHTPP, prior to each stress session. During WS, estrogen signaling through ERβ was responsible for the behavioral sensitization to repeated social stress. Sucrose preference, acoustic startle, and marble burying tasks determined that blocking ERβ in the CeA during WS prevented the development of depressive-, anxiety-like, and hypervigilant behaviors. Additionally, brain analysis revealed a long-term decrease of intra-CeA CRF expression in PHTPP-treated rats. These experiments indicate that ERβ signaling in the CeA, likely through its effects on CRF, contributes to the development of negative valence behaviors that result from exposure to repeated social stress in female rats.
Collapse
Key Words
- ACTH, adrenocorticotropic hormone
- ASR, acoustic startle responding
- Anxiety
- BCA, bicinchoninic acid
- CON, control handing
- CORT, corticosterone
- CRF, corticotropin releasing factor
- CeA, central amygdala
- Central amygdala
- Corticotropin releasing factor
- EPM, elevated plus maze
- ERβ, estrogen receptor beta
- Estrogen receptor beta
- HPA, hypothalamic pituitary adrenal axis
- LC, locus coeruleus
- MB, marble burying
- PHTPP, 4-[2-Phenyl-5: 7-bis (trifluoromethyl) pyrazolo [1,5-a] pyrimidine-3- yl] phenol
- SPT, sucrose preference testing
- Social stress
- WS, witness stress
- dB, decibels
Collapse
Affiliation(s)
- Cora E. Smiley
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
- Dorn VA Medical Center, Columbia, SC, USA
| | - Brittany S. Pate
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
- University of South Carolina, Arnold School of Public Health, Department of Exercise Science, Columbia, SC, USA
| | - Samantha J. Bouknight
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Megan J. Francis
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Alexandria V. Nowicki
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Evelynn N. Harrington
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Susan K. Wood
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
- Dorn VA Medical Center, Columbia, SC, USA
| |
Collapse
|
12
|
Frankfurt M, Nassrallah Z, Luine V. Steroid Hormone Interaction with Dendritic Spines: Implications for Neuropsychiatric Disease. ADVANCES IN NEUROBIOLOGY 2023; 34:349-366. [PMID: 37962800 DOI: 10.1007/978-3-031-36159-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dendritic spines, key sites for neural plasticity, are influenced by gonadal steroids. In this chapter, we review the effects of gonadal steroids on dendritic spine density in areas important to cognitive function, the hippocampus, and prefrontal cortex. Most of these animal model studies investigated the effects of estrogen in females, but we also include more recent data on androgen effects in both males and females. The underlying genomic and non-genomic mechanisms related to gonadal steroid-induced spinogenesis are also reviewed. Subsequently, we discuss possible reasons for the observed sex differences in many neuropsychiatric diseases, which appear to be caused, in part, by aberrant synaptic connections that may involve dendritic spine pathology. Overall, knowledge concerning the regulation of dendritic spines by gonadal hormones has grown since the initial discoveries in the 1990s, and current research points to a potential role for aberrant spine functioning in many neuropsychiatric disorders.
Collapse
Affiliation(s)
- Maya Frankfurt
- Hofstra Northwell School of Nursing and Physician Assistant Studies, Hempstead, NY, USA.
| | - Zeinab Nassrallah
- Department of Science Education Zucker School of Medicine, 500 Hofstra University, Hempstead, NY, USA
| | - Victoria Luine
- Department of Psychology, Hunter College, New York, NY, USA
| |
Collapse
|
13
|
Larosa A, Wong TP. The hippocampus in stress susceptibility and resilience: Reviewing molecular and functional markers. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110601. [PMID: 35842073 DOI: 10.1016/j.pnpbp.2022.110601] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/22/2022] [Accepted: 07/10/2022] [Indexed: 10/17/2022]
Abstract
Understanding the individual variability that comes with the likelihood of developing stress-related psychopathologies is of paramount importance when addressing mechanisms of their neurobiology. This article focuses on the hippocampus as a region that is highly influenced by chronic stress exposure and that has strong ties to the development of related disorders, such as depression and post-traumatic stress disorder. We first outline three commonly used animal models that have been used to separate animals into susceptible and resilient cohorts. Next, we review molecular and functional hippocampal markers of susceptibility and resilience. We propose that the hippocampus plays a crucial role in the differences in the processing and storage of stress-related information in animals with different stress susceptibilities. These hippocampal markers not only help us attain a more comprehensive understanding of the various facets of stress-related pathophysiology, but also could be targeted for the development of new treatments.
Collapse
Affiliation(s)
- Amanda Larosa
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Tak Pan Wong
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada; Dept. of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
14
|
Gut Microsex/Genderome, Immunity and the Stress Response in the Sexes: An Updated Review. SEXES 2022. [DOI: 10.3390/sexes3040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Sex has been universally acknowledged as a confounding factor in every type of biological study, while there are strong sex differences in morbidity along the lifespan. Humans have almost identical genomes (99.2%), yet minor variance in their DNA produces remarkable phenotypic diversity across the human population. On the other hand, metagenomic analysis of the human microbiome is more variable, depending on the sex, lifestyle, geography, and age of individuals under study. Immune responses in humans also exhibit variations, with an especially striking sexual dimorphism, which is at play in several other physiologic processes. Sex steroids have noticeable effects on the composition of the human microbiome along the lifespan, accompanied by parallel changes in immunity and the stress response. Gut microsex/genderome, a recently coined term, defines the sexually dimorphic gut microbiome. Apart from the sex steroids, the stress hormones are also at play in the proliferation of microbes. This review summarizes the concept of gut microsex/genderome under the prism of recent studies on the interrelations of the sexually dimorphic microbiome with immunity and stress.
Collapse
|
15
|
Dong TN, Kramár EA, Beardwood JH, Al-Shammari A, Wood MA, Keiser AA. Temporal endurance of exercise-induced benefits on hippocampus-dependent memory and synaptic plasticity in female mice. Neurobiol Learn Mem 2022; 194:107658. [PMID: 35811066 PMCID: PMC9901197 DOI: 10.1016/j.nlm.2022.107658] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/20/2022] [Accepted: 07/04/2022] [Indexed: 02/08/2023]
Abstract
Exercise facilitates hippocampal neurogenesis and neuroplasticity that in turn, promotes cognitive function. Our previous studies have demonstrated that in male mice, voluntary exercise enables hippocampus-dependent learning in conditions that are normally subthreshold for long-term memory formation in sedentary animals. Such cognitive enhancement can be maintained long after exercise has ceased and can be re-engaged by a subsequent subthreshold exercise session, suggesting exercise-induced benefits are temporally dynamic. In females, the extent to which the benefits of exercise can be maintained and the mechanisms underlying this maintenance have yet to be defined. Here, we examined the exercise parameters required to initiate and maintain the benefits of exercise in female C57BL/6J mice. Using a subthreshold version of the hippocampus-dependent task called object-location memory (OLM) task, we show that 14d of voluntary exercise enables learning under subthreshold acquisition conditions in female mice. Following the initial exercise, a 7d sedentary delay results in diminished performance, which can be re-facilitated when animals receive 2d of reactivating exercise following the sedentary delay. Assessment of estrous cycle reveals enhanced wheel running activity during the estrus phase relative to the diestrus phase, whereas estrous phase on training or test had no effect on OLM performance. Utilizing the same exercise parameters, we demonstrate that 14d of exercise enhances long-term potentiation (LTP) in the CA1 region of the hippocampus, an effect that persists throughout the sedentary delay and following the reactivating exercise session. Previous studies have proposed exercise-induced BDNF upregulation as the mechanism underlying exercise-mediated benefits on synaptic plasticity and cognition. However, our assessment of hippocampal Bdnf mRNA expression following memory retrieval reveals no difference between exercise conditions and control, suggesting that persistent Bdnf upregulation may not be required for maintenance of exercise-induced benefits. Together, our data indicate that 14d of voluntary exercise can initiate long-lasting benefits on neuroplasticity and cognitive function in female mice, establishing the first evidence on the temporal endurance of exercise-induced benefits in females.
Collapse
Affiliation(s)
- T N Dong
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - E A Kramár
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - J H Beardwood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - A Al-Shammari
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - M A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - A A Keiser
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States.
| |
Collapse
|
16
|
Hokenson RE, Alam YH, Short AK, Jung S, Jang C, Baram TZ. Sex-dependent effects of multiple acute concurrent stresses on memory: a role for hippocampal estrogens. Front Behav Neurosci 2022; 16:984494. [PMID: 36160685 PMCID: PMC9492881 DOI: 10.3389/fnbeh.2022.984494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022] Open
Abstract
Memory disruption commonly follows chronic stress, whereas acute stressors are generally benign. However, acute traumas such as mass shootings or natural disasters—lasting minutes to hours and consisting of simultaneous physical, social, and emotional stresses—are increasingly recognized as significant risk factors for memory problems and PTSD. Our prior work has revealed that these complex stresses (concurrent multiple acute stresses: MAS) disrupt hippocampus-dependent memory in male rodents. In females, the impacts of MAS are estrous cycle-dependent: MAS impairs memory during early proestrus (high estrogens phase), whereas the memory of female mice stressed during estrus (low estrogens phase) is protected. Female memory impairments limited to high estrogens phases suggest that higher levels of estrogens are necessary for MAS to disrupt memory, supported by evidence that males have higher hippocampal estradiol than estrous females. To test the role of estrogens in stress-induced memory deficits, we blocked estrogen production using aromatase inhibitors. A week of blockade protected male and female mice from MAS-induced memory disturbances, suggesting that high levels of estrogens are required for stress-provoked memory impairments in both males and females. To directly quantify 17β-estradiol in murine hippocampus we employed both ELISA and mass spectrometry and identified significant confounders in both procedures. Taken together, the cross-cycle and aromatase studies in males and females support the role for high hippocampal estrogens in mediating the effect of complex acute stress on memory. Future studies focus on the receptors involved, the longevity of these effects, and their relation to PTSD-like behaviors in experimental models.
Collapse
Affiliation(s)
- Rachael E. Hokenson
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, United States
- *Correspondence: Rachael E. Hokenson
| | - Yasmine H. Alam
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Annabel K. Short
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA =, United States
| | - Sunhee Jung
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Tallie Z. Baram
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA =, United States
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
17
|
Bowman R, Frankfurt M, Luine V. Sex differences in cognition following variations in endocrine status. Learn Mem 2022; 29:234-245. [PMID: 36206395 PMCID: PMC9488023 DOI: 10.1101/lm.053509.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/03/2022] [Indexed: 11/24/2022]
Abstract
Spatial memory, mediated primarily by the hippocampus, is responsible for orientation in space and retrieval of information regarding location of objects and places in an animal's environment. Since the hippocampus is dense with steroid hormone receptors and is capable of robust neuroplasticity, it is not surprising that changes in spatial memory performance occur following a variety of endocrine alterations. Here, we review cognitive changes in both spatial and nonspatial memory tasks following manipulations of the hypothalamic-pituitary-adrenal and gonadal axes and after exposure to endocrine disruptors in rodents. Chronic stress impairs male performance on numerous behavioral cognitive tasks and enhances or does not impact female cognitive function. Sex-dependent changes in cognition following stress are influenced by both organizational and activational effects of estrogen and vary depending on the developmental age of the stress exposure, but responses to gonadal hormones in adulthood are more similar than different in the sexes. Also discussed are possible underlying neural mechanisms for these steroid hormone-dependent, cognitive effects. Bisphenol A (BPA), an endocrine disruptor, given at low levels during adolescent development, impairs spatial memory in adolescent male and female rats and object recognition memory in adulthood. BPA's negative effects on memory may be mediated through alterations in dendritic spine density in areas that mediate these cognitive tasks. In summary, this review discusses the evidence that endocrine status of an animal (presence or absence of stress hormones, gonadal hormones, or endocrine disruptors) impacts cognitive function and, at times, in a sex-specific manner.
Collapse
Affiliation(s)
- Rachel Bowman
- Department of Psychology, Sacred Heart University, Fairfield, Connecticut 06825, USA
| | - Maya Frankfurt
- Department of Psychology, Sacred Heart University, Fairfield, Connecticut 06825, USA
- Hofstra Northwell School of Nursing and Physician Assistant Studies, Hofstra University, Hempstead, New York 11549, USA
| | - Victoria Luine
- Department of Psychology, Hunter College of City University of New York, New York, New York 10065, USA
| |
Collapse
|
18
|
Cathomas F, Holt LM, Parise EM, Liu J, Murrough JW, Casaccia P, Nestler EJ, Russo SJ. Beyond the neuron: Role of non-neuronal cells in stress disorders. Neuron 2022; 110:1116-1138. [PMID: 35182484 PMCID: PMC8989648 DOI: 10.1016/j.neuron.2022.01.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
Abstract
Stress disorders are leading causes of disease burden in the U.S. and worldwide, yet available therapies are fully effective in less than half of all individuals with these disorders. Although to date, much of the focus has been on neuron-intrinsic mechanisms, emerging evidence suggests that chronic stress can affect a wide range of cell types in the brain and periphery, which are linked to maladaptive behavioral outcomes. Here, we synthesize emerging literature and discuss mechanisms of how non-neuronal cells in limbic regions of brain interface at synapses, the neurovascular unit, and other sites of intercellular communication to mediate the deleterious, or adaptive (i.e., pro-resilient), effects of chronic stress in rodent models and in human stress-related disorders. We believe that such an approach may one day allow us to adopt a holistic "whole body" approach to stress disorder research, which could lead to more precise diagnostic tests and personalized treatment strategies. Stress is a major risk factor for many psychiatric disorders. Cathomas et al. review new insight into how non-neuronal cells mediate the deleterious effects, as well as the adaptive, protective effects, of stress in rodent models and human stress-related disorders.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leanne M Holt
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - James W Murrough
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrizia Casaccia
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
19
|
Czarzasta K, Bogacki-Rychlik W, Segiet-Swiecicka A, Kruszewska J, Malik J, Skital V, Kasarello K, Wrzesien R, Bialy M, Sajdel-Sulkowska EM. Gender differences in short- vs. long-term impact of maternal depression following pre-gestational chronic mild stress. Exp Neurol 2022; 353:114059. [DOI: 10.1016/j.expneurol.2022.114059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 03/24/2022] [Accepted: 03/27/2022] [Indexed: 11/04/2022]
|
20
|
Short AK, Thai CW, Chen Y, Kamei N, Pham AL, Birnie MT, Bolton JL, Mortazavi A, Baram TZ. Single-Cell Transcriptional Changes in Hypothalamic Corticotropin-Releasing Factor-Expressing Neurons After Early-Life Adversity Inform Enduring Alterations in Vulnerabilities to Stress. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 3:99-109. [PMID: 36712559 PMCID: PMC9874075 DOI: 10.1016/j.bpsgos.2021.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/11/2021] [Accepted: 12/03/2021] [Indexed: 02/01/2023] Open
Abstract
Background Mental health and vulnerabilities to neuropsychiatric disorders involve the interplay of genes and environment, particularly during sensitive developmental periods. Early-life adversity (ELA) and stress promote vulnerabilities to stress-related affective disorders, yet it is unknown how transient ELA dictates lifelong neuroendocrine and behavioral reactions to stress. The population of hypothalamic corticotropin-releasing factor (CRF)-expressing neurons that regulate stress responses is a promising candidate to mediate the long-lasting influences of ELA on stress-related behavioral and hormonal responses via enduring transcriptional and epigenetic mechanisms. Methods Capitalizing on a well-characterized model of ELA, we examined ELA-induced changes in gene expression profiles of CRF-expressing neurons in the hypothalamic paraventricular nucleus of developing male mice. We used single-cell RNA sequencing on isolated CRF-expressing neurons. We determined the enduring functional consequences of transcriptional changes on stress reactivity in adult ELA mice, including hormonal responses to acute stress, adrenal weights as a measure of chronic stress, and behaviors in the looming shadow threat task. Results Single-cell transcriptomics identified distinct and novel CRF-expressing neuronal populations, characterized by both their gene expression repertoire and their neurotransmitter profiles. ELA-provoked expression changes were selective to specific subpopulations and affected genes involved in neuronal differentiation, synapse formation, energy metabolism, and cellular responses to stress and injury. Importantly, these expression changes were impactful, apparent from adrenal hypertrophy and augmented behavioral responses to stress in adulthood. Conclusions We uncover a novel repertoire of stress-regulating CRF cell types differentially affected by ELA and resulting in augmented stress vulnerability, with relevance to the origins of stress-related affective disorders.
Collapse
Affiliation(s)
- Annabel K. Short
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Christina W. Thai
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California
| | - Yuncai Chen
- Department of Pediatrics, University of California Irvine, Irvine, California
| | - Noriko Kamei
- Department of Pediatrics, University of California Irvine, Irvine, California
| | - Aidan L. Pham
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Matthew T. Birnie
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Jessica L. Bolton
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California
| | - Tallie Z. Baram
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California,Department of Pediatrics, University of California Irvine, Irvine, California,Department of Neurology, University of California Irvine, Irvine, California,Address correspondence to Tallie Z. Baram, M.D., Ph.D.
| |
Collapse
|
21
|
Bhatnagar S. Rethinking stress resilience. Trends Neurosci 2021; 44:936-945. [PMID: 34711401 DOI: 10.1016/j.tins.2021.09.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/04/2021] [Accepted: 09/30/2021] [Indexed: 01/10/2023]
Abstract
Resilience to stressful life events has received considerable attention in both clinical and preclinical studies. A number of neural substrates have been identified as putatively mediating resilience to stress. However, there remains considerable diversity in how resilience is defined and studied. This article aims to examine how resilience is defined and conceptualized in social psychology, public health, and related fields, to better inform the understanding of stress resilience in the neurobiological context, and to differentiate resilience from other patterns of response to stressful experiences. An understanding of resilience through the lens of clinical and applied sciences is likely to lead to the identification of more robust and reproducible neural substrates, though many challenges remain.
Collapse
Affiliation(s)
- Seema Bhatnagar
- Stress Neurobiology Center, Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia Research Institute, The Perelman School of Medicine at the University of Pennsylvania, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA.
| |
Collapse
|
22
|
Godó S, Barabás K, Lengyel F, Ernszt D, Kovács T, Kecskés M, Varga C, Jánosi TZ, Makkai G, Kovács G, Orsolits B, Fujiwara T, Kusumi A, Ábrahám IM. Single-Molecule Imaging Reveals Rapid Estradiol Action on the Surface Movement of AMPA Receptors in Live Neurons. Front Cell Dev Biol 2021; 9:708715. [PMID: 34631701 PMCID: PMC8495425 DOI: 10.3389/fcell.2021.708715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023] Open
Abstract
Gonadal steroid 17β-estradiol (E2) exerts rapid, non-genomic effects on neurons and strictly regulates learning and memory through altering glutamatergic neurotransmission and synaptic plasticity. However, its non-genomic effects on AMPARs are not well understood. Here, we analyzed the rapid effect of E2 on AMPARs using single-molecule tracking and super-resolution imaging techniques. We found that E2 rapidly decreased the surface movement of AMPAR via membrane G protein-coupled estrogen receptor 1 (GPER1) in neurites in a dose-dependent manner. The cortical actin network played a pivotal role in the GPER1 mediated effects of E2 on the surface mobility of AMPAR. E2 also decreased the surface movement of AMPAR both in synaptic and extrasynaptic regions on neurites and increased the synaptic dwell time of AMPARs. Our results provide evidence for understanding E2 action on neuronal plasticity and glutamatergic neurotransmission at the molecular level.
Collapse
Affiliation(s)
- Soma Godó
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Klaudia Barabás
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Ferenc Lengyel
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Dávid Ernszt
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Tamás Kovács
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Miklós Kecskés
- PTE-NAP Cortical Microcircuits Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, Pécs, Hungary
| | - Csaba Varga
- PTE-NAP Cortical Microcircuits Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, Pécs, Hungary
| | - Tibor Z Jánosi
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Géza Makkai
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Gergely Kovács
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Barbara Orsolits
- Laboratory of Neuroimmunology, Institute of Experimental Medicine of the Hungarian Academy of Sciences, Budapest, Hungary
| | - Takahiro Fujiwara
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
| | - Akihiro Kusumi
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Onna, Japan
| | - István M Ábrahám
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| |
Collapse
|
23
|
Palamarchuk IS, Vaillancourt T. Mental Resilience and Coping With Stress: A Comprehensive, Multi-level Model of Cognitive Processing, Decision Making, and Behavior. Front Behav Neurosci 2021; 15:719674. [PMID: 34421556 PMCID: PMC8377204 DOI: 10.3389/fnbeh.2021.719674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/14/2021] [Indexed: 11/13/2022] Open
Abstract
Aversive events can evoke strong emotions that trigger cerebral neuroactivity to facilitate behavioral and cognitive shifts to secure physiological stability. However, upon intense and/or chronic exposure to such events, the neural coping processes can be maladaptive and disrupt mental well-being. This maladaptation denotes a pivotal point when psychological stress occurs, which can trigger subconscious, "automatic" neuroreactivity as a defence mechanism to protect the individual from potential danger including overwhelming unpleasant feelings and disturbing or threatening thoughts.The outcomes of maladaptive neural activity are cognitive dysfunctions such as altered memory, decision making, and behavior that impose a risk for mental disorders. Although the neurocognitive phenomena associated with psychological stress are well documented, the complex neural activity and pathways related to stressor detection and stress coping have not been outlined in detail. Accordingly, we define acute and chronic stress-induced pathways, phases, and stages in relation to novel/unpredicted, uncontrollable, and ambiguous stressors. We offer a comprehensive model of the stress-induced alterations associated with multifaceted pathophysiology related to cognitive appraisal and executive functioning in stress.
Collapse
Affiliation(s)
- Iryna S Palamarchuk
- Counselling Psychology, Faculty of Education, University of Ottawa, Ottawa, ON, Canada
| | - Tracy Vaillancourt
- Counselling Psychology, Faculty of Education, University of Ottawa, Ottawa, ON, Canada.,School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
24
|
Wu Q, Wang B, Ntim M, Zhang X, Na XY, Yuan YH, Wu XF, Yang JY, Li S. SRC-1 Deficiency Increases Susceptibility of Mice to Depressive-Like Behavior After Exposure to CUMS. Neurochem Res 2021; 46:1830-1843. [PMID: 33881662 DOI: 10.1007/s11064-021-03316-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 11/24/2022]
Abstract
Steroid receptor coactivator 1 (SRC-1) is one of the coactivators recruited by the nuclear receptors (NRs) when NRs are activated by steroid hormones, such as glucocorticoid. SRC-1 is abundant in hippocampus and hypothalamus and is also related to some major risk factors for depression, implicated by its reduced expression after stress and its effect on hypothalamus-pituitary-adrenal gland axis function. However, whether SRC-1 is involved in the formation of depression remains unclear. In this study, we firstly established chronic unpredictable stress (CUS) to induce depressive-like behaviors in mice and found that SRC-1 expression was reduced by CUS. A large number of studies have shown that neuroinflammation is associated with stress-induced depression and lipopolysaccharide (LPS) injection can lead to neuroinflammation and depressive-like behaviors in mice. Our result indicated that LPS treatment also decreased SRC-1 expression in mouse brain, implying the involvement of SRC-1 in the process of inflammation and depression. Next, we showed that the chronic unpredictable mild stress (CUMS) failed to elicit the depressive-like behaviors and dramatically promoted the expression of SRC-1 in brain of wild type mice. What's more, the SRC-1 knockout mice were more susceptible to CUMS to develop depressive-like behaviors and presented the changed expression of glucocorticoid receptor. However, SRC-1 deficiency did not affect the microglia activation induced by CUMS. Altogether, these results indicate a correlation between SRC-1 level and depressive-like behaviors, suggesting that SRC-1 might be involved in the development of depression induced by stress.
Collapse
Affiliation(s)
- Qiong Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Bin Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Michael Ntim
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xuan Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Xue-Yan Na
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yu-Hui Yuan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xue-Fei Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| | - Jin-Yi Yang
- Department of Urology, Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, China.
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| |
Collapse
|
25
|
Florido A, Velasco ER, Soto-Faguás CM, Gomez-Gomez A, Perez-Caballero L, Molina P, Nadal R, Pozo OJ, Saura CA, Andero R. Sex differences in fear memory consolidation via Tac2 signaling in mice. Nat Commun 2021; 12:2496. [PMID: 33941789 PMCID: PMC8093426 DOI: 10.1038/s41467-021-22911-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 03/25/2021] [Indexed: 11/08/2022] Open
Abstract
Memory formation is key for brain functioning. Uncovering the memory mechanisms is helping us to better understand neural processes in health and disease. Moreover, more specific treatments for fear-related disorders such as posttraumatic stress disorder and phobias may help to decrease their negative impact on mental health. In this line, the Tachykinin 2 (Tac2) pathway in the central amygdala (CeA) has been shown to be sufficient and necessary for the modulation of fear memory consolidation. CeA-Tac2 antagonism and its pharmacogenetic temporal inhibition impair fear memory in male mice. Surprisingly, we demonstrate here the opposite effect of Tac2 blockade on enhancing fear memory consolidation in females. Furthermore, we show that CeA-testosterone in males, CeA-estradiol in females and Akt/GSK3β/β-Catenin signaling both mediate the opposite-sex differential Tac2 pathway regulation of fear memory.
Collapse
Affiliation(s)
- A Florido
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - E R Velasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - C M Soto-Faguás
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - A Gomez-Gomez
- Integrative Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - L Perez-Caballero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - P Molina
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Unitat de Fisiologia Animal, Departament de Biologia Cel·lular, Fisiologia i Immunologia. Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - R Nadal
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Centro de Investigación Biomédica En Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - O J Pozo
- Integrative Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - C A Saura
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - R Andero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
- Centro de Investigación Biomédica En Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
- ICREA, Pg. Lluís Companys 23, Barcelona, Spain.
| |
Collapse
|
26
|
Khaleghi M, Rajizadeh MA, Bashiri H, Kohlmeier KA, Mohammadi F, Khaksari M, Shabani M. Estrogen attenuates physical and psychological stress-induced cognitive impairments in ovariectomized rats. Brain Behav 2021; 11:e02139. [PMID: 33811472 PMCID: PMC8119870 DOI: 10.1002/brb3.2139] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/14/2021] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Women are more vulnerable to stress-related disorders than men, which is counterintuitive as female sex hormones, especially estrogen, have been shown to be protective against stress disorders. METHODS In this study, we investigated whether two different models of stress act differently on ovariectomized (OVX) rats and the impact of estrogen on physical or psychological stress-induced impairments in cognitive-behaviors. Adult female Wistar rats at 21-22 weeks of age were utilized for this investigation. Sham and OVX rats were subjected to physical and psychological stress for 1 hr/day for 7 days, and cognitive performance was assessed using morris water maze (MWM) and passive avoidance (PA) tests. The open field and elevated plus maze tests (EPM) evaluated exploratory and anxiety-like behaviors. RESULTS In sham and OVX rats, both physical and psychological stressors were associated with an increase in EPM-determined anxiety-like behavior. OVX rats exhibited decreased explorative behavior in comparison with nonstressed sham rats (p < .05). Both physical stress and psychological stress resulted in disrupted spatial cognition as assayed in the MWM (p < .05) and impaired learning and memory as determined by the PA test when the OVX and sham groups were compared with the nonstressed sham group. Estrogen increased explorative behavior, learning and memory (p < .05), and decreased anxiety-like behavior compared with vehicle in OVX rats exposed to either type of stressor. CONCLUSIONS When taken together, estrogen and both stressors had opposite effects on memory, anxiety, and PA performance in a rat model of menopause, which has important implications for potential protective effects of estrogen in postmenopausal women exposed to chronic stress.
Collapse
Affiliation(s)
- Mina Khaleghi
- Department of Physiology and Pharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Rajizadeh
- Department of Physiology and Pharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Department of Physiology and Pharmacology, Kerman University of Medical Science, Kerman Iran and Sirjan School of Medical Sciences, Sirjan, Iran
| | - Kristi Anne Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fatemeh Mohammadi
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|