1
|
Pereira AAR, Pinto AM, Malerba HN, Toricelli M, Buck HS, Viel TA. Microdose lithium improves behavioral deficits and modulates molecular mechanisms of memory formation in female SAMP-8, a mouse model of accelerated aging. PLoS One 2024; 19:e0299534. [PMID: 38574297 PMCID: PMC10994667 DOI: 10.1371/journal.pone.0299534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/12/2024] [Indexed: 04/06/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neuronal disorder that leads to the development of dementia. Until nowadays, some therapies may alleviate the symptoms, but there is no pharmacological treatment. Microdosing lithium has been used to modify the pathological characteristics of the disease, with effects in both experimental and clinical conditions. The present work aimed to analyze the effects of this treatment on spatial memory, anxiety, and molecular mechanisms related to long-term memory formation during the aging process of a mouse model of accelerated aging (SAMP-8). Female SAMP-8 showed learning and memory impairments together with disruption of memory mechanisms, neuronal loss, and increased density of senile plaques compared to their natural control strain, the senescence-accelerated mouse resistant (SAMR-1). Chronic treatment with lithium promoted memory maintenance, reduction in anxiety, and maintenance of proteins related to memory formation and neuronal density. The density of senile plaques was also reduced. An increase in the density of gamma-aminobutyric acid A (GABAA) and α7 nicotinic cholinergic receptors was also observed and related to neuroprotection and anxiety reduction. In addition, this microdose of lithium inhibited the activation of glycogen synthase kinase-3beta (GSK-3β), the classical mechanism of lithium cell effects, which could contribute to the preservation of the memory mechanism and reduction in senile plaque formation. This work shows that lithium effects in neuroprotection along the aging process are not related to a unique cellular mechanism but produce multiple effects that slowly protect the brain along the aging process.
Collapse
Affiliation(s)
- Arthur Antonio Ruiz Pereira
- Department of Pharmacology, Institute of Biomedical Sciences, Graduate Course on Pharmacology, Universidade de Sao Paulo, Sao Paulo, Sao Paulo, Brazil
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Alessandra Macedo Pinto
- Graduate Course on Gerontology, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Helena Nascimento Malerba
- Department of Pharmacology, Institute of Biomedical Sciences, Graduate Course on Pharmacology, Universidade de Sao Paulo, Sao Paulo, Sao Paulo, Brazil
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Mariana Toricelli
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Hudson Sousa Buck
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
- Department of Physiology, University of Mogi das Cruzes, Mogi das Cruzes, Sao Paulo, Brazil
| | - Tania Araujo Viel
- Department of Pharmacology, Institute of Biomedical Sciences, Graduate Course on Pharmacology, Universidade de Sao Paulo, Sao Paulo, Sao Paulo, Brazil
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
- Graduate Course on Gerontology, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
2
|
Knouse MC, Deutschmann AU, Nenov MN, Wimmer ME, Briand LA. Sex differences in pre- and post-synaptic glutamate signaling in the nucleus accumbens core. Biol Sex Differ 2023; 14:52. [PMID: 37596655 PMCID: PMC10439632 DOI: 10.1186/s13293-023-00537-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/08/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Glutamate signaling within the nucleus accumbens underlies motivated behavior and is involved in psychiatric disease. Although behavioral sex differences in these processes are well-established, the neural mechanisms driving these differences are largely unexplored. In these studies, we examine potential sex differences in synaptic plasticity and excitatory transmission within the nucleus accumbens core. Further understanding of baseline sex differences in reward circuitry will shed light on potential mechanisms driving behavioral differences in motivated behavior and psychiatric disease. METHODS Behaviorally naïve adult male and female Long-Evans rats, C57Bl/6J mice, and constitutive PKMζ knockout mice were killed and tissue containing the nucleus accumbens core was collected for ex vivo slice electrophysiology experiments. Electrophysiology recordings examined baseline sex differences in synaptic plasticity and transmission within this region and the potential role of PKMζ in long-term depression. RESULTS Within the nucleus accumbens core, both female mice and rats exhibit higher AMPA/NMDA ratios compared to male animals. Further, female mice have a larger readily releasable pool of glutamate and lower release probability compared to male mice. No significant sex differences were detected in spontaneous excitatory postsynaptic current amplitude or frequency. Finally, the threshold for induction of long-term depression was lower for male animals than females, an effect that appears to be mediated, in part, by PKMζ. CONCLUSIONS We conclude that there are baseline sex differences in synaptic plasticity and excitatory transmission in the nucleus accumbens core. Our data suggest there are sex differences at multiple levels in this region that should be considered in the development of pharmacotherapies to treat psychiatric illnesses such as depression and substance use disorder.
Collapse
Affiliation(s)
- Melissa C Knouse
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Andre U Deutschmann
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Miroslav N Nenov
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Mathieu E Wimmer
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Lisa A Briand
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA.
- Neuroscience Program, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA.
| |
Collapse
|
3
|
Schecter RW, Jensen CM, Gavornik JP. Sex and estrous cycle affect experience-dependent plasticity in mouse primary visual cortex. PLoS One 2023; 18:e0282349. [PMID: 37068089 PMCID: PMC10109517 DOI: 10.1371/journal.pone.0282349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/13/2023] [Indexed: 04/18/2023] Open
Abstract
Sex hormones can affect cellular physiology and modulate synaptic plasticity, but it is not always clear whether or how sex-dependent differences identified in vitro express themselves as functional dimorphisms in the brain. Historically, most experimental neuroscience has been conducted using only male animals and the literature is largely mute about whether including female mice in will introduce variability due to inherent sex differences or endogenous estrous cycles. Though this is beginning to change following an NIH directive that sex should be included as a factor in vertebrate research, the lack of information raises practical issues around how to design experimental controls and apply existing knowledge to more heterogeneous populations. Various lines of research suggest that visual processing can be affected by sex and estrous cycle stage. For these reasons, we performed a series of in vivo electrophysiological experiments to characterize baseline visual function and experience-dependent plasticity in the primary visual cortex (V1) of male and female mice. We find that sex and estrous stage have no statistically significant effect on baseline acuity measurements, but that both sex and estrous stage have can modulate two mechanistically distinct forms of experience dependent cortical plasticity. We also demonstrate that resulting variability can be largely controlled with appropriate normalizations. These findings suggest that V1 plasticity can be used for mechanistic studies focusing on how sex hormones effect experience dependent plasticity in the mammalian cortex.
Collapse
Affiliation(s)
- Rachel W. Schecter
- Center for Systems Neuroscience, Biology Department, Boston University, Boston, MA, United States of America
| | - Cambria M. Jensen
- Center for Systems Neuroscience, Biology Department, Boston University, Boston, MA, United States of America
| | - Jeffrey P. Gavornik
- Center for Systems Neuroscience, Biology Department, Boston University, Boston, MA, United States of America
| |
Collapse
|
4
|
Fox LC, Scholl JL, Watt MJ, Forster GL. GABA A Receptor and Serotonin Transporter Expression Changes Dissociate Following Mild Traumatic Brain Injury: Influence of Sex and Estrus Cycle Phase in Rats. Neuroscience 2023; 514:38-55. [PMID: 36736883 DOI: 10.1016/j.neuroscience.2023.01.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
Mild traumatic brain injuries (mild TBIs) can affect both males and females, but females are more likely to report long-term psychological complications, including changes in mood and generalized anxiety. Additionally, reproductive cycle phase has been shown to affect mild TBI symptom expression within females. These variances may result from sex differences in mild TBI-induced alterations to neurotransmission in brain regions that influence mood and emotion, possibly mediated by sex steroids. The hippocampus and amygdala are implicated in stress responses and anxiety, and within these regions, gamma-aminobutyric acid (GABA) and serotonin modulate output and behavioral expression. Metabolites of progesterone can allosterically enhance GABAergic signaling, and sex steroids are suggested to regulate the expression of the serotonin transporter (SERT). To determine how mild TBI might alter GABA receptor and SERT expression in males and females, immunocytochemistry was used to quantify expression of the alpha-1 subunit of the GABAA receptor (α1-GABAA), SERT, and a neuronal marker (NeuN) in the brains of adult male and naturally-cycling female rats, both with and without mild TBI, 17 days after injury. Mild TBI altered the expression of α1-GABAA in the amygdala and hippocampus in both sexes, but the direction of change observed depended on sex and reproductive cycle phase. In contrast, mild TBI had little effect on SERT expression. However, SERT expression differed between sexes and varied with the cycle phase. These findings demonstrate that regulation of neurotransmission following mild TBI differs between males and females, with implications for behavioral outcomes and the efficacy of therapeutic strategies.
Collapse
Affiliation(s)
- Laura C Fox
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark St, Vermillion, SD, USA.
| | - Jamie L Scholl
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark St, Vermillion, SD, USA.
| | - Michael J Watt
- Center for Brain and Behavior Research, Department of Anatomy, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| | - Gina L Forster
- Center for Brain and Behavior Research, Department of Anatomy, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| |
Collapse
|
5
|
Towers EB, Kilgore M, Bakhti-Suroosh A, Pidaparthi L, Williams IL, Abel JM, Lynch WJ. Sex differences in the neuroadaptations associated with incubated cocaine-craving: A focus on the dorsomedial prefrontal cortex. Front Behav Neurosci 2023; 16:1027310. [PMID: 36688133 PMCID: PMC9854116 DOI: 10.3389/fnbeh.2022.1027310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Women have a shorter course from initial cocaine use to meeting the criteria for cocaine use disorder as compared to men. Preclinical findings similarly indicate that females develop key features of an addiction-like phenotype faster than males, including an enhanced motivation for cocaine and compulsive use, indicating that this phenomenon is biologically based. The goals of this study were to determine whether cocaine-craving, another key feature of addiction, also develops sooner during withdrawal in females than males and to determine whether there are sex differences in the molecular mechanisms associated with its development focusing on markers known to mediate cocaine-craving in males (i.e., dorsomedial prefrontal cortex, dmPFC, expression of brain-derived neurotrophic factor exon-IV, Bdnf-IV, and NMDA receptor subunits, Grin2a, Grin2b, and Grin1). Methods Cocaine-craving was assessed following extended-access cocaine self-administration and 2, 7, or 14 days of withdrawal using an extinction/cue-induced reinstatement procedure. Tissue was obtained from the dmPFC immediately after reinstatement testing and gene expression changes were analyzed using real-time qPCR. Results In males, cocaine-craving (total extinction and cue-induced reinstatement responding) progressively increased from early to later withdrawal time-points whereas in females, cocaine-craving was already elevated during early withdrawal (after 2 days) and did not further increase at later withdrawal time-points. Levels of cocaine-craving, however, were similar between the sexes. Gene expression changes differed markedly between the sexes such that males showed the expected relapse- and withdrawal-associated changes in Bdnf-IV, Grin2a, Grin2b, and Grin1 expression, but females only showed a modest increase Grin1 expression at the intermediate withdrawal timepoint. Discussion These findings indicate that cocaine-craving is similarly expressed in males and females although the time-course for its incubation appears to be accelerated in females; the molecular mechanisms also likely differ in females versus males.
Collapse
Affiliation(s)
- Eleanor Blair Towers
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
- Medical Scientist Training Program, University of Virginia, Charlottesville, VA, United States
| | - Madison Kilgore
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| | - Anousheh Bakhti-Suroosh
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| | - Lasyapriya Pidaparthi
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| | - Ivy L. Williams
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| | - Jean M. Abel
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| | - Wendy J. Lynch
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
6
|
Hosseinzadeh Sahafi O, Rezayof A, Ghasemzadeh Z, Alijanpour S, Rahimian S. Ameliorating effect offluoxetine on tamoxifen-induced memory loss: The role of corticolimbic NMDA receptors and CREB/BDNF/cFos signaling pathways in rats. Brain Res 2022; 1794:148058. [PMID: 36007581 DOI: 10.1016/j.brainres.2022.148058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/02/2022]
Abstract
Tamoxifen-induced cognitive dysfunction may lead to fluoxetine consumption in patients with breast cancer. Since the brain mechanisms are unclear in tamoxifen/fluoxetine therapy, the blockade effect of hippocampal/amygdala/prefrontal cortical NMDA receptors was examined in fluoxetine/tamoxifen-induced memory retrieval. We also assessed the corticolimbic signaling pathways in memory retrieval under the drug treatment in adult male Wistar rats. Using the Western blot technique, the expression levels of the cAMP response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF), and cFos were evaluated in the corticolimbic regions. The results showed that pre-test administration of fluoxetine (3 and 5 mg/kg, i.p.) improved tamoxifen-induced memory impairment in the passive avoidance learning task. Pre-test bilateral microinjection of D-AP5, a selective NMDA receptor antagonist, into the dorsal hippocampal CA1 regions and the central amygdala (CeA), but not the medial prefrontal cortex (mPFC), inhibited the improving effect of fluoxetine on tamoxifen response. It is important to note that the microinjection of D-AP5 into the different sites by itself did not affect memory retrieval. Memory retrieval increased the signaling pathway of pCREB/CREB/BDNF/cFos in the corticolimbic regions. Tamoxifen-induced memory impairment decreased the hippocampal/PFC BDNF level and the amygdala level of pCREB/CREB/cFos. The improving effect of fluoxetine on tamoxifen significantly increased the hippocampal/PFC expression levels of BDNF, the PFC/amygdala expression levels of cFos, and the ratio of pCREB/CREB in all targeted areas. Thus, NMDA receptors' activity in the different corticolimbic regions mediates fluoxetine/tamoxifen memory retrieval. The corticolimbic synaptic plasticity changes likely accompany the improving effect of fluoxetine on tamoxifen response.
Collapse
Affiliation(s)
- Oveis Hosseinzadeh Sahafi
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Zahra Ghasemzadeh
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Sakineh Alijanpour
- Department of Biology, Faculty of Science, Gonbad Kavous University, Gonbad Kavous, Iran
| | - Sepehrdad Rahimian
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
7
|
Chimeric Structures in Mental Illnesses-"Magic" Molecules Specified for Complex Disorders. Int J Mol Sci 2022; 23:ijms23073739. [PMID: 35409098 PMCID: PMC8998808 DOI: 10.3390/ijms23073739] [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: 03/13/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Mental health problems cover a wide spectrum of diseases, including mild to moderate anxiety, depression, alcohol/drug use disorders, as well as bipolar disorder and schizophrenia. Pharmacological treatment seems to be one of the most effective opportunities to recover function efficiently and satisfactorily. However, such disorders are complex as several target points are involved. This results in a necessity to combine different types of drugs to obtain the necessary therapeutic goals. There is a need to develop safer and more effective drugs. Considering that mental illnesses share multifactorial processes, the paradigm of one treatment with multiple modes of action rather than single-target strategies would be more effective for successful therapies. Therefore, hybrid molecules that combine two pharmacophores in one entity show promise, as they possess the desired therapeutic index with a small off-target risk. This review aims to provide information on chimeric structures designed for mental disorder therapy (i.e., schizophrenia and depression), and new types of drug candidates currently being tested. In addition, a discussion on some benefits and limitations of multifunctional, bivalent drug candidates is also given.
Collapse
|
8
|
Zhang HL, Zhao B, Yang P, Du YQ, Han W, Xu J, Yin DM. Steroid Receptor Coactivator 3 Regulates Synaptic Plasticity and Hippocampus-dependent Memory. Neurosci Bull 2021; 37:1645-1657. [PMID: 34228315 PMCID: PMC8643392 DOI: 10.1007/s12264-021-00741-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/27/2021] [Indexed: 11/28/2022] Open
Abstract
Steroid hormones play important roles in brain development and function. The signaling of steroid hormones depends on the interaction between steroid receptors and their coactivators. Although the function of steroid receptor coactivators has been extensively studied in other tissues, their functions in the central nervous system are less well investigated. In this study, we addressed the function of steroid receptor coactivator 3 (SRC3) - a member of the p160 SRC protein family that is expressed predominantly in the hippocampus. While hippocampal development was not altered in Src3+/- mice, hippocampus-dependent functions such as short-term memory and spatial memory were impaired. We further demonstrated that the deficient learning and memory in Src3+/- mice was strongly associated with the impairment of long-term potentiation (LTP) at Schaffer Collateral-CA1 synapses. Mechanistic studies indicated that Src3+/- mutation altered the composition of N-methyl-D-aspartate receptor subunits in the postsynaptic densities of hippocampal neurons. Finally, we showed that SRC3 regulated synaptic plasticity and learning mainly dependent on its lysine acetyltransferase activity. Taken together, these results reveal previously unknown functions of SRC3 in the hippocampus and thus may provide insight into how steroid hormones regulate brain function.
Collapse
Affiliation(s)
- Hai-Long Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Bing Zhao
- MOE Frontiers Center for Brain Science, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Pin Yang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Yin-Quan Du
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Wei Han
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dong-Min Yin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
9
|
Knouse MC, Briand LA. Behavioral sex differences in cocaine and opioid use disorders: The role of gonadal hormones. Neurosci Biobehav Rev 2021; 128:358-366. [PMID: 34214512 DOI: 10.1016/j.neubiorev.2021.06.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/20/2021] [Accepted: 06/27/2021] [Indexed: 11/20/2022]
Abstract
Females are more vulnerable than males to many aspects of cocaine use disorder. This vulnerability also translates to opioid use disorder, with females exhibiting stronger behavioral responses than males to drugs such as heroin and morphine. While there is evidence for many overlapping neural mechanisms underlying cocaine and opioid abuse, there is also a breadth of evidence indicating divergent effects of the drugs on synaptic plasticity. This makes it unclear whether the behavioral sex differences seen in substance use disorder across different drugs of abuse rely on the same mechanisms. Ovarian hormones have consistently been implicated as drivers of the behavioral sex differences in cocaine taking and seeking. While there are far fewer studies on the role of ovarian hormones in opioid use disorder, the existing data suggest that ovarian hormones may not drive these behavioral effects in the same manner as in cocaine use disorder. This review highlights evidence that behavioral sex differences in substance use disorder might be driven by different mechanisms depending on drug class.
Collapse
Affiliation(s)
| | - Lisa A Briand
- Department of Psychology, Temple University, United States; Neuroscience Program, Temple University, United States.
| |
Collapse
|
10
|
Abstract
Migraine is considered mostly a woman’s complaint, even if it affects also men. Epidemiological data show a higher incidence of the disease in women, starting from puberty throughout life. The sex-related differences of migraine hold clinical relevance too. The frequency, duration, and disability of attacks tend to be higher in women. Because of this, probably, they also consult specialists more frequently and take more prescription drugs than men. Different mechanisms have been evaluated to explain these differences. Hormonal milieu and its modulation of neuronal and vascular reactivity is probably one of the most important aspects. Estrogens and progesterone regulate a host of biological functions through two mechanisms: nongenomic and genomic. They influence several neuromediators and neurotransmitters, and they may cause functional and structural differences in several brain regions, involved in migraine pathogenesis. In addition to their central action, sex hormones exert rapid modulation of vascular tone. The resulting specific sex phenotype should be considered during clinical management and experimental studies.
Collapse
|
11
|
Dean B, Gogos A. The impact of ovariectomy and chronic estrogen treatment on gene expression in the rat cortex: Implications for psychiatric disorders. Psychoneuroendocrinology 2021; 127:105192. [PMID: 33730612 DOI: 10.1016/j.psyneuen.2021.105192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 01/01/2023]
Abstract
Estrogens, via estrogen-mediated changes in CNS function, have been suggested to be beneficial in the treatment of several psychiatric disorders. Few studies have used transcriptomic technologies to determine the effect of estrogen on gene expression in the CNS. Thus, we aimed to examine the impact of ovariectomy (the removal of all ovarian hormones) and estrogen replacement on rat frontal cortical gene expression. We used the Agilent SurePrint G3 Gene Expression Rat Array to measure levels of RNA in intact (cycling) female rats and in ovariectomized rats that were, or were not, given 17β-estradiol in implants for 4 weeks. Compared to untreated ovariectomized rats, intact rats (effect of ovarian hormones; comparison 1) and rats receiving 17β-estradiol replacement (estrogen-specific effects; comparison 2) showed significant changes in cortical gene expression (58 and 36 genes, respectively). These changes in gene expression would be expected to affect pathways that regulate neurotransmitters, glutathione and sphingolipids; pathways known to be implicated in the pathophysiologies of psychiatric disorders. When we compared the levels of gene expression in the two comparisons that had a significance of p < 0.01 independent of magnitude of change, there was a strong correlation between fold changes in gene expression for 127 genes. We posit that this correlation is due to the level of expression of these genes being strongly influenced by both cycling and replacement estrogen. Further exploration of ovarian hormone- and estrogen-sensitive gene expression may provide new insight into the aetiology of aspects of psychiatric disorders that show sex differences.
Collapse
Affiliation(s)
- Brian Dean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
12
|
Xu YH, Wang XX, Wang MJ, Liu YY, Xue Z, Chen JX. Influence of progestational stress on BDNF and NMDARs in the hippocampus of male offspring and amelioration by Chaihu Shugan San. Biomed Pharmacother 2021; 135:111204. [PMID: 33548869 DOI: 10.1016/j.biopha.2020.111204] [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: 08/11/2020] [Revised: 12/13/2020] [Accepted: 12/26/2020] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Progestational stress has been proven to be a risk for the neural development of offspring, especially in the hippocampus. However, whether Chaihu Shugan San (CSS) can ameliorate hippocampal neural development via the regulation of brain-derived neurotrophic factor (BDNF), and N-methyl-D-aspartate receptors (NMDAR) 2A (NR2A) and 2B (NR2B), and the mechanism of such action remains unclear. METHODS Thirty-six female rats were randomly allocated into control, chronic immobilization stress (CIS) and CSS groups according to the random number table, respectively. The male offspring were fed for 21 days after birth then randomly divided into the same three groups (6 rats/group) as the female rats. Female rats, except for the control group, underwent 21-day CIS to established a progestational stress anxiety-like model which was evaluated by body weight, the elevated plus-maze (EPM) test and serum dopamine (DA) measured using an enzyme-linked immunosorbent assay (ELISA). The expression levels of estrogen receptors (ERα/ERβ) and progesterone receptor (PR) in female rat ovaries were quantified by real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. The hippocampal tissue in the 21-day offspring was observed by hematoxylin-eosin (HE) staining. The concentration of BDNF, NR2A, and NR2B were measured by RT-qPCR and immunohistochemistry in the CA3 and dentate gyrus (DG) regions of offsprings' hippocampus. RESULTS Compared with the female control group, significant differences in body weight, EPM test and DA concentration were observed in the CIS group, meanwhile, the concentration of ERα (P < 0.05), PR (P < 0.05) and ERβ in the ovaries were decreased. In the offsprings' hippocampus of the CIS group, the chromatin of the nucleus was edge set and with condensed and irregular morphology nucleus, and the cytoplasm was unevenly stained with spaces around the cells, moreover, the expression levels of BDNF, NR2A, and NR2B were also declined (P < 0.05). However, Chaihu Shugan San reversed these changes, especially the BDNF in the DG region (P < 0.05), and NR2A and NR2B in the CA3 and DG region (P < 0.05). CONCLUSIONS CSS could ameliorate the neural development of the hippocampus in offspring damaged by anxiety-like progestational stress in female rats via regulating the expression levels of ERα, ERβ, and PR in female rat ovaries and BDNF, NR2A, and NR2B in the hippocampus of their offspring.
Collapse
MESH Headings
- Animals
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Disease Models, Animal
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Estrogen Receptor beta/genetics
- Estrogen Receptor beta/metabolism
- Female
- Gestational Age
- Hippocampus/drug effects
- Hippocampus/metabolism
- Hippocampus/pathology
- Male
- Neurogenesis/drug effects
- Ovary/drug effects
- Ovary/metabolism
- Plant Extracts/pharmacology
- Pregnancy
- Prenatal Exposure Delayed Effects
- Rats, Wistar
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Restraint, Physical
- Signal Transduction
- Stress, Psychological/drug therapy
- Stress, Psychological/genetics
- Stress, Psychological/metabolism
- Stress, Psychological/pathology
- Rats
Collapse
Affiliation(s)
- Ya-Hui Xu
- School of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin-Xing Wang
- School of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Ming-Jing Wang
- School of Basic Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Yue-Yun Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhe Xue
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
13
|
Rapid effects of neurosteroids on neuronal plasticity and their physiological and pathological implications. Neurosci Lett 2021; 750:135771. [PMID: 33636284 DOI: 10.1016/j.neulet.2021.135771] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 11/22/2022]
Abstract
Current neuroscience research on neurosteroids and their synthetic analogues - neuroactive steroids - clearly demonstrate their drug likeness in a variety of neurological and psychiatric conditions. Moreover, research on neurosteroids continues to provide novel mechanistic insights into receptor activation or inhibition of various receptors. This mini-review will provide a high-level overview of the research area and discuss the various classes of potential physiological and pathological implications discovered so far.
Collapse
|
14
|
Liu Y, Wang Z, Zhang X, Li S, Wu W, Li X, Yang Y. A sex-dependent delayed maturation of visual plasticity induced by adverse experiences in early childhood. Neurobiol Stress 2020; 13:100256. [PMID: 33344711 PMCID: PMC7739182 DOI: 10.1016/j.ynstr.2020.100256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/04/2020] [Accepted: 10/08/2020] [Indexed: 11/27/2022] Open
Abstract
Adverse experiences in early life have a long-term impact on the development of brain, which in turn increases the susceptibility to mental illness during adulthood, especially in female subjects. However, whether and how the visual cortex is affected by these adverse experiences as well as the mechanisms underlying the sex difference are largely unknown. Here, we established a new mouse model of early-life chronic mild stress (ECMS) without anxiety or depression-like behavior in adulthood. ECMS mice showed normal maturation of visual acuity and orientation/direction selectivity, whereas their visual cortical neurons preferred lower spatial frequency (SF) and higher temporal frequency (TF) than control mice. Meanwhile the development of ocular dominance (OD) plasticity was delayed. Specifically, compared with control mice, ECMS mice in the early stage of the critical period (CP) showed a reduction in GABA synthesis enzyme expression as well as lower OD plasticity which could be occluded by diazepam. In contrast, ECMS mice in the late stage of CP showed stronger OD plasticity, accompanied by higher expression of N-methyl-D-aspartate (NMDA) receptor NR2B subunit. Interestingly, only female ECMS mice at adulthood maintained juvenile-like OD plasticity as well as high NR2B expressions. Artificial increase in estradiol level in ECMS males via estradiol supplementary diminished this sex difference. Lastly, OD plasticity was abolished in adult ECMS females either performed with the bilateral ovariectomy in prepuberty, or directly infused with NR2B antagonist Ro 25–6981 into the visual cortex. Overall, our study demonstrates that early adverse experiences have a lasting effect on visual development of mice in a sex-dependent manner, which is mediated by the estradiol-NR2B pathway.
Collapse
Affiliation(s)
- Yueqin Liu
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zhenni Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xinxin Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Sitong Li
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Wei Wu
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xin Li
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yupeng Yang
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| |
Collapse
|
15
|
Tozzi A, Bellingacci L, Pettorossi VE. Rapid Estrogenic and Androgenic Neurosteroids Effects in the Induction of Long-Term Synaptic Changes: Implication for Early Memory Formation. Front Neurosci 2020; 14:572511. [PMID: 33192257 PMCID: PMC7653679 DOI: 10.3389/fnins.2020.572511] [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: 06/14/2020] [Accepted: 08/21/2020] [Indexed: 11/17/2022] Open
Abstract
Mounting experimental evidence demonstrate that sex neuroactive steroids (neurosteroids) are essential for memory formation. Neurosteroids have a profound impact on the function and structure of neural circuits and their local synthesis is necessary for the induction of both long-term potentiation (LTP) and long-term depression (LTD) of synaptic transmission and for neural spine formation in different areas of the central nervous system (CNS). Several studies demonstrated that in the hippocampus, 17β-estradiol (E2) is necessary for inducing LTP, while 5α-dihydrotestosterone (DHT) is necessary for inducing LTD. This contribution has been proven by administering sex neurosteroids in rodent models and by using blocking agents of their synthesis or of their specific receptors. The general opposite role of sex neurosteroids in synaptic plasticity appears to be dependent on their different local availability in response to low or high frequency of synaptic stimulation, allowing the induction of bidirectional synaptic plasticity. The relevant contribution of these neurosteroids to synaptic plasticity has also been described in other brain regions involved in memory processes such as motor learning, as in the case of the vestibular nuclei, the cerebellum, and the basal ganglia, or as the emotional circuit of the amygdala. The rapid effects of sex neurosteroids on neural synaptic plasticity need the maintenance of a tonic or phasic local steroid synthesis determined by neural activity but might also be influenced by circulating hormones, age, and gender. To disclose the exact mechanisms how sex neurosteroids participate in finely tuning long-term synaptic changes and spine remodeling, further investigation is required.
Collapse
Affiliation(s)
- Alessandro Tozzi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Laura Bellingacci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | |
Collapse
|
16
|
McDougall SA, Apodaca MG, Park GI, Teran A, Baum TJ, Montejano NR. MK801-induced locomotor activity in preweanling and adolescent male and female rats: role of the dopamine and serotonin systems. Psychopharmacology (Berl) 2020; 237:2469-2483. [PMID: 32445054 PMCID: PMC7354898 DOI: 10.1007/s00213-020-05547-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 05/05/2020] [Indexed: 01/01/2023]
Abstract
RATIONALE MK801, like other NMDA receptor open-channel blockers (e.g., ketamine and phencyclidine), increases the locomotor activity of rats and mice. Whether this behavioral effect ultimately relies on monoamine neurotransmission is of dispute. OBJECTIVE The purpose of this study was to determine whether these psychopharmacological effects and underlying neural mechanisms vary according to sex and age. METHODS Across four experiments, male and female preweanling and adolescent rats were pretreated with vehicle, the monoamine-depleting agent reserpine (1 or 5 mg/kg), the dopamine (DA) synthesis inhibitor ∝-methyl-DL-p-tyrosine (AMPT), the serotonin (5-HT) synthesis inhibitor 4-chloro-DL-phenylalanine methyl ester hydrochloride (PCPA), or both AMPT and PCPA. The locomotor activity of preweanling and adolescent rats was then measured after saline or MK801 (0.3 mg/kg) treatment. RESULTS As expected, MK801 increased the locomotor activity of all age groups and both sexes, but the stimulatory effects were significantly less pronounced in male adolescent rats. Preweanling rats and adolescent female rats were more sensitive to the effects of DA and 5-HT synthesis inhibitors, as AMPT and PCPA caused only small reductions in the MK801-induced locomotor activity of male adolescent rats. Co-administration of AMPT+PCPA or high-dose reserpine (5 mg/kg) treatment substantially reduced MK801-induced locomotor activity in both age groups and across both sexes. CONCLUSIONS These results, when combined with other recent studies, show that NMDA receptor open-channel blockers cause pronounced age-dependent behavioral effects that can vary according to sex. The neural changes underlying these sex and age differences appear to involve monoamine neurotransmission.
Collapse
Affiliation(s)
- Sanders A McDougall
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA.
| | - Matthew G Apodaca
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| | - Ginny I Park
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| | - Angie Teran
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| | - Timothy J Baum
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| | - Nazaret R Montejano
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA, 92407, USA
| |
Collapse
|
17
|
Nicholson K, MacLusky NJ, Leranth C. Synaptic effects of estrogen. VITAMINS AND HORMONES 2020; 114:167-210. [PMID: 32723543 DOI: 10.1016/bs.vh.2020.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The concept that estradiol may act as a local neuromodulator in the brain, rapidly affecting connectivity and synaptic function, has been firmly established by research over the last 30 years. De novo synthesis of estradiol within the brain as well as signaling mechanisms mediating responses to the hormone have been demonstrated, along with morphological evidence indicating rapid changes in synaptic input following increases in local estradiol levels. These rapid synaptic effects may play important roles in both physiological and pathophysiological responses to changes in circulating hormone levels, as well as in neurodegenerative disease. How local effects of estradiol on synaptic plasticity are integrated into changes in the overall activity of neural networks in the brain, however, remains a subject that is only incompletely understood.
Collapse
Affiliation(s)
- Kate Nicholson
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Neil J MacLusky
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Csaba Leranth
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, School of Medicine, New Haven, CT, United States.
| |
Collapse
|
18
|
Moghimi P, Jimenez NT, McLoon LK, Netoff TI, Lee MS, MacDonald A, Miller RF. Electoretinographic evidence of retinal ganglion cell-dependent function in schizophrenia. Schizophr Res 2020; 219:34-46. [PMID: 31615740 PMCID: PMC7442157 DOI: 10.1016/j.schres.2019.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 02/02/2023]
Abstract
Schizophrenia is a complex disorder that is diagnosed mainly with clinical observation and evaluation. Recent studies suggest that many people with schizophrenia have abnormalities in the function of the N-methyl-d-aspartate receptor (NMDAR). The retina is part of the central nervous system and expresses the NMDAR, raising the possibility of the early detection of NMDAR-related schizophrenia by detecting differences in retinal function. As a first-step, we used two non-invasive outpatient tests of retinal function, the photopic negative response (PhNR) of the light-adapted flash-electroretinogram (PhNR-fERG) and the pattern ERG (PERG), to test individuals with schizophrenia and controls to determine if there were measurable differences between the two populations. The PhNR-fERG showed that males with schizophrenia had a significant increase in the variability of the overall response, which was not seen in the females with schizophrenia. Additionally at the brightest flash strength, there were significant increases in the PhNR amplitude in people with schizophrenia that were maximal in controls. Our results show measurable dysfunction of retinal ganglion cells (RGCs) in schizophrenia using the PhNR-fERG, with a good deal of variability in the retinal responses of people with schizophrenia. The PhNR-fERG holds promise as a method to identify individuals more at risk for developing schizophrenia, and may help understand heterogeneity in etiology and response to treatment.
Collapse
Affiliation(s)
- Pantea Moghimi
- Department of Neurobiology, University of Chicago, Chicago, IL, United States of America
| | - Nathalia Torres Jimenez
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States of America,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America,Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States of America
| | - Linda K. McLoon
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States of America,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America,Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States of America
| | - Theoden I. Netoff
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States of America,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America,Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Michael S. Lee
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States of America
| | - Angus MacDonald
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States of America; Departments of Psychology and Psychiatry, University of Minnesota, Minneapolis, MN, United States of America.
| | - Robert F. Miller
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States of America,Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America,Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, United States of America
| |
Collapse
|
19
|
Paredes S, Cantillo S, Candido KD, Knezevic NN. An Association of Serotonin with Pain Disorders and Its Modulation by Estrogens. Int J Mol Sci 2019; 20:E5729. [PMID: 31731606 PMCID: PMC6888666 DOI: 10.3390/ijms20225729] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
Ovarian hormones play an important role in pain perception, and are responsible, at least in part, for the pain threshold differences between the sexes. Modulation of pain and its perception are mediated by neurochemical changes in several pathways, affecting both the central and peripheral nervous systems. One of the most studied neurotransmitters related to pain disorders is serotonin. Estrogen can modify serotonin synthesis and metabolism, promoting a general increase in its tonic effects. Studies evaluating the relationship between serotonin and disorders such as irritable bowel syndrome, fibromyalgia, migraine, and other types of headache suggest a clear impact of this neurotransmitter, thereby increasing the interest in serotonin as a possible future therapeutic target. This literature review describes the importance of substances such as serotonin and ovarian hormones in pain perception and illustrates the relationship between those two, and their direct influence on the presentation of the aforementioned pain-related conditions. Additionally, we review the pathways and receptors implicated in each disorder. Finally, the objective was to stimulate future pharmacological research to experimentally evaluate the potential of serotonin modulators and ovarian hormones as therapeutic agents to regulate pain in specific subpopulations.
Collapse
Affiliation(s)
- Stephania Paredes
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, 836 W. Wellington Ave. Suite 4815, Chicago, IL 60657, USA; (S.P.); (S.C.); (K.D.C.)
| | - Santiago Cantillo
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, 836 W. Wellington Ave. Suite 4815, Chicago, IL 60657, USA; (S.P.); (S.C.); (K.D.C.)
| | - Kenneth D. Candido
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, 836 W. Wellington Ave. Suite 4815, Chicago, IL 60657, USA; (S.P.); (S.C.); (K.D.C.)
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
- Department of Surgery, University of Illinois, Chicago, IL 60612, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, 836 W. Wellington Ave. Suite 4815, Chicago, IL 60657, USA; (S.P.); (S.C.); (K.D.C.)
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
- Department of Surgery, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
20
|
Ovalles AC, Contoreggi NH, Marques-Lopes J, Van Kempen TA, Iadecola C, Waters EM, Glass MJ, Milner TA. Plasma Membrane Affiliated AMPA GluA1 in Estrogen Receptor β-containing Paraventricular Hypothalamic Neurons Increases Following Hypertension in a Mouse Model of Post-menopause. Neuroscience 2019; 423:192-205. [PMID: 31682817 DOI: 10.1016/j.neuroscience.2019.09.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
Sex and ovarian function contribute to hypertension susceptibility, however, the mechanisms are not well understood. Prior studies show that estrogens and neurogenic factors, including hypothalamic glutamatergic NMDA receptor plasticity, play significant roles in rodent hypertension. Here, we investigated the role of sex and ovarian failure on AMPA receptor plasticity in estrogen-sensitive paraventricular nucleus (PVN) neurons in naïve and angiotensin II (AngII) infused male and female mice and female mice at early and late stages of accelerated ovarian failure (AOF). High-resolution electron microscopy was used to assess the subcellular distribution of AMPA GluA1 in age-matched male and female estrogen receptor beta (ERβ) enhanced green fluorescent protein (EGFP) reporter mice as well as female ERβ-EGFP mice treated with 4-vinylcyclohexene diepoxide. In the absence of AngII, female mice at a late stage of AOF displayed higher levels of GluA1 on the plasma membrane, indicative of functional protein, in ERβ-expressing PVN dendrites when compared to male, naïve female and early stage AOF mice. Following slow-pressor AngII infusion, males, as well as early and late stage AOF females had elevated blood pressure. Significantly, only late stage-AOF female mice infused with AngII had an increase in GluA1 near the plasma membrane in dendrites of ERβ-expressing PVN neurons. In contrast, prior studies reported that plasmalemmal NMDA GluN1 increased in ERβ-expressing PVN dendrites in males and early, but not late stage AOF females. Together, these findings reveal that early and late stage AOF female mice display unique molecular signatures of long-lasting synaptic strength prior to, and following hypertension.
Collapse
Affiliation(s)
- Astrid C Ovalles
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Tracey A Van Kempen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Michael J Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA.
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
21
|
Alonso-Caraballo Y, Ferrario CR. Effects of the estrous cycle and ovarian hormones on cue-triggered motivation and intrinsic excitability of medium spiny neurons in the Nucleus Accumbens core of female rats. Horm Behav 2019; 116:104583. [PMID: 31454509 PMCID: PMC7256930 DOI: 10.1016/j.yhbeh.2019.104583] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 11/08/2022]
Abstract
Naturally occurring alterations in estradiol influence food intake in females. However, how motivational responses to food cues are affected by the estrous cycle or ovarian hormones is unknown. In addition, while individual susceptibility to obesity is accompanied by enhanced incentive motivational responses to food cues and increased NAc intrinsic excitability in males, studies in females are absent. Therefore, we examined basal differences in intrinsic NAc excitability of obesity-prone vs. obesity-resistant females and determined how conditioned approach (a measure of cue-triggered motivation), food intake, and motivation for food vary with the cycle in naturally cycling female obesity-prone, obesity-resistant, and outbred Sprague-Dawley rats. Finally, we used ovariectomy followed by hormone treatment to determine the role of ovarian hormones in cue-triggered motivation in selectively-bred and outbred female rats. We found that intrinsic excitability of NAc MSNs and conditioned approach are enhanced in female obesity-prone vs. obesity-resistant rats. These effects were driven by greater MSN excitability and conditioned approach behavior during metestrus/diestrus vs. proestrus/estrus in obesity-prone but not obesity-resistant rats, despite similar regulation of food intake and food motivation by the cycle in these groups. Furthermore, estradiol and progesterone treatment reduced conditioned approach behavior in obesity-prone and outbred Sprague-Dawley females. To our knowledge, these data are the first to demonstrate cycle- and hormone-dependent effects on the motivational response to a food cue, and the only studies to date to determine how individual susceptibility to obesity influences NAc excitability, cue-triggered food-seeking, and differences in the regulation of these neurobehavioral responses by the estrous cycle.
Collapse
Affiliation(s)
| | - Carrie R Ferrario
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States of America; Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States of America.
| |
Collapse
|
22
|
McDougall SA, Park GI, Ramirez GI, Gomez V, Adame BC, Crawford CA. Sex-dependent changes in ketamine-induced locomotor activity and ketamine pharmacokinetics in preweanling, adolescent, and adult rats. Eur Neuropsychopharmacol 2019; 29:740-755. [PMID: 30981586 PMCID: PMC7059997 DOI: 10.1016/j.euroneuro.2019.03.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/22/2019] [Accepted: 03/27/2019] [Indexed: 01/06/2023]
Abstract
Although ketamine has long been known to increase locomotor activity, only recently was it realized that this behavioral effect varies according to both sex and age. The purpose of the present study was threefold: first, to measure the locomotor activating effects of ketamine in male and female rats across early ontogeny and into adulthood; second, to assess ketamine and norketamine pharmacokinetics in the dorsal striatum and hippocampus of the same age groups; and, third, to use curvilinear regression to determine the relationship between locomotor activity and dorsal striatal concentrations of ketamine and norketamine. A high dose of ketamine (80 mg/kg, i.p.) was administered in order to examine the complete cycle of locomotor responsiveness across a 280-min testing session. In separate groups of rats, the dorsal striata and hippocampi were removed at 10 time points (0-360 min) after ketamine administration and samples were assayed for ketamine, norketamine, and dopamine using HPLC. In female rats, ketamine produced high levels of locomotor activity that varied only slightly among age groups. Male preweanling rats responded like females, but adolescent and adult male rats exhibited lesser amounts of ketamine-induced locomotor activity. Ketamine and norketamine pharmacokinetics, especially peak values and area under the curve, generally mirrored age- and sex-dependent differences in locomotor activity. Among male rats and younger female rats, dorsal striatal ketamine and norketamine levels accounted for a large proportion of the variance in locomotor activity. In adult female rats, however, an additional factor, perhaps involving other ketamine and norketamine metabolites, was influencing locomotor activity.
Collapse
Affiliation(s)
- Sanders A McDougall
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA.
| | - Ginny I Park
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| | - Goretti I Ramirez
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| | - Vanessa Gomez
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| | - Brittnee C Adame
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| | - Cynthia A Crawford
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| |
Collapse
|
23
|
Role of Steroid Therapy after Ischemic Stroke by n-Methyl-d-Aspartate Receptor Gene Regulation. J Stroke Cerebrovasc Dis 2018; 27:3066-3075. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.06.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/14/2018] [Accepted: 06/30/2018] [Indexed: 12/13/2022] Open
|
24
|
Grigsby KB, Kovarik CM, Rottinghaus GE, Booth FW. High and low nightly running behavior associates with nucleus accumbens N-Methyl-d-aspartate receptor (NMDAR) NR1 subunit expression and NMDAR functional differences. Neurosci Lett 2018; 671:50-55. [PMID: 29425730 DOI: 10.1016/j.neulet.2018.02.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/03/2018] [Accepted: 02/05/2018] [Indexed: 11/19/2022]
Abstract
The extent to which N-Methyl-d-aspartate (NMDA) receptors facilitate the motivation to voluntarily wheel-run in rodents has yet to be determined. In so, we utilized female Wistar rats selectively bred to voluntarily run high (HVR) and low (LVR) nightly distances in order to examine if endogenous differences in nucleus accumbens (NAc) NMDA receptor expression and function underlies the propensity for high or low motivation to voluntarily wheel-run. 12-14 week old HVR and LVR females were used to examine: 1.) NAc mRNA and protein expression of NMDA subunits NR1, NR2A and NR2B; 2.) NMDA current responses in isolated medium spiny neurons (MSNs) and 3.) NMDA-evoked dopamine release in an ex vivo preparation of NAc punches. Expectedly, there was a large divergence in nightly running distance and time between HVR and LVR rats. We saw a significantly higher mRNA and protein expression of NR1 in HVR compared to LVR rats, while seeing no difference in the expression of NR2A or NR2B. There was a greater current response to a 500 ms application of 300 μM of NMDA in medium-spiny neurons isolated from the NAc HVR compared to LVR animals. On average, NMDA-evoked punches (50 μM of NMDA for 10 min) taken from HVR rats retained ∼54% of the dopamine content compared to their bilateral non-evoked sides, while evoked punches from LVR animals showed no statistical decrease in dopamine content compared to their non-evoked sides. Collectively, these data suggest a potential link between NAc NR1 subunit expression as well as NMDA function and the predisposition for nightly voluntary running behavior in rats. In light of the epidemic rise in physical inactivity, these findings have the potential to explain a neuro-molecular mechanism that regulates the motivation to be physically active.
Collapse
Affiliation(s)
- Kolter B Grigsby
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States.
| | - Cathleen M Kovarik
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - George E Rottinghaus
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States; Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
25
|
McCarthny CR, Du X, Wu YC, Hill RA. Investigating the Interactive Effects of Sex Steroid Hormones and Brain-Derived Neurotrophic Factor during Adolescence on Hippocampal NMDA Receptor Expression. Int J Endocrinol 2018; 2018:7231915. [PMID: 29666640 PMCID: PMC5831834 DOI: 10.1155/2018/7231915] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/05/2017] [Accepted: 10/19/2017] [Indexed: 12/18/2022] Open
Abstract
Sex steroid hormones have neuroprotective properties which may be mediated by brain-derived neurotrophic factor (BDNF). This study sought to determine the interactive effects of preadolescent hormone manipulation and BDNF heterozygosity (+/-) on hippocampal NMDA-R expression. Wild-type and BDNF+/- mice were gonadectomised, and females received either 17β-estradiol or progesterone treatment, while males received either testosterone or dihydrotestosterone (DHT) treatment. Dorsal (DHP) and ventral hippocampus (VHP) were dissected, and protein expression of GluN1, GluN2A, GluN2B, and PSD-95 was assessed by Western blot analysis. Significant genotype × OVX interactions were found for GluN1 and GluN2 expression within the DHP of female mice, suggesting modulation of select NMDA-R levels by female sex hormones is mediated by BDNF. Furthermore, within the DHP BDNF+/- mice show a hypersensitive response to hormone treatment on GluN2 expression which may result from upstream alterations in TrkB phosphorylation. In contrast to the DHP, the VHP showed no effects of hormone manipulation but significant effects of genotype on NMDA-R expression. Castration had no effect on NMDA-R expression; however, androgen treatment had selective effects on GluN2B. These data show case distinct, interactive roles for sex steroid hormones and BDNF in the regulation of NMDA-R expression that are dependent on dorsal versus ventral hippocampal region.
Collapse
Affiliation(s)
- Cushla R. McCarthny
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | - Xin Du
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - YeeWen Candace Wu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rachel A. Hill
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
26
|
Pogun S, Yararbas G, Nesil T, Kanit L. Sex differences in nicotine preference. J Neurosci Res 2017; 95:148-162. [PMID: 27870459 DOI: 10.1002/jnr.23858] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/29/2016] [Accepted: 07/11/2016] [Indexed: 01/01/2023]
Abstract
Smoking is the major cause of preventable deaths worldwide, and although there is a decline in overall smoking prevalence in developed countries, the decline in women is less pronounced than in men. Women become dependent faster and experience greater difficulties in quitting. Similar trends have been observed in animal models of nicotine/tobacco addiction. Individual differences in vulnerability to drug abuse are also observed in nicotine/tobacco addiction and point to the importance of sex differences. This Review, summarizes findings from three experimental approaches used to depict nicotine preference in animal models, intravenous and oral nicotine self-administration and nicotine-induced conditioned place preference. Nicotine preference is considered to be reflected in the animal's motivation to administer the drug (intravenously or orally) or to prefer an environment paired with the presence of the drug (conditioned place preference). These approaches all point to the importance of sex and age of the subjects; the preference of females and adolescents appear to be more pronounced than that of males and adults, respectively. A closer look at these factors will help us understand the mechanisms that underlie nicotine addiction and develop strategies to cope. Ignoring sex differences and reaching conclusions based only on studies using male subjects has resulted in erroneous generalizations in the past. Sex differences in nicotine preference have been clearly documented, and awareness on this aspect of nicotine dependence will significantly impact our success in translational research. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sakire Pogun
- Center for Brain Research, Ege University, Izmir, Turkey
| | - Gorkem Yararbas
- Institute on Drug Abuse, Toxicology and Pharmaceutical Science, Ege University, Izmir, Turkey
| | - Tanseli Nesil
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia
| | - Lutfiye Kanit
- Center for Brain Research, Ege University, Izmir, Turkey.,Physiology Department, School of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
27
|
van den Buuse M, Low JK, Kwek P, Martin S, Gogos A. Selective enhancement of NMDA receptor-mediated locomotor hyperactivity by male sex hormones in mice. Psychopharmacology (Berl) 2017; 234:2727-2735. [PMID: 28674745 DOI: 10.1007/s00213-017-4668-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 05/29/2017] [Indexed: 01/03/2023]
Abstract
RATIONALE Altered glutamate NMDA receptor function is implicated in schizophrenia, and gender differences have been demonstrated in this illness. OBJECTIVES This study aimed to investigate the interaction of gonadal hormones with NMDA receptor-mediated locomotor hyperactivity and PPI disruption in mice. RESULTS The effect of 0.25 mg/kg of MK-801 on locomotor activity was greater in male mice than in female mice. Gonadectomy (by surgical castration) significantly reduced MK-801-induced hyperlocomotion in male mice, but no effect of gonadectomy was seen in female mice or on amphetamine-induced locomotor hyperactivity. The effect of MK-801 on prepulse inhibition of startle (PPI) was similar in intact and castrated male mice and in ovariectomized (OVX) female mice. In contrast, there was no effect of MK-801 on PPI in intact female mice. Forebrain NMDA receptor density, as measured with [3H]MK-801 autoradiography, was significantly higher in male than in female mice but was not significantly altered by either castration or OVX. CONCLUSIONS These results suggest that male sex hormones enhance the effect of NMDA receptor blockade on psychosis-like behaviour. This interaction was not seen in female mice and was independent of NMDA receptor density in the forebrain. Male sex hormones may be involved in psychosis by an interaction with NMDA receptor hypofunction.
Collapse
Affiliation(s)
- Maarten van den Buuse
- Mental Health Research Institute, Parkville, VIC, Australia. .,Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia. .,The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia. .,School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia.
| | - Jac Kee Low
- Mental Health Research Institute, Parkville, VIC, Australia.,School of Nursing and Midwifery, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Perrin Kwek
- Mental Health Research Institute, Parkville, VIC, Australia
| | - Sally Martin
- Mental Health Research Institute, Parkville, VIC, Australia
| | - Andrea Gogos
- Mental Health Research Institute, Parkville, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
28
|
Estradiol and raloxifene modulate hippocampal gamma oscillations during a spatial memory task. Psychoneuroendocrinology 2017; 78:85-92. [PMID: 28183030 DOI: 10.1016/j.psyneuen.2017.01.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/16/2017] [Accepted: 01/22/2017] [Indexed: 11/22/2022]
Abstract
Previous work suggests that estradiol regulates the expression of hippocampal parvalbumin as well as hippocampus-dependent spatial memory in mice. Parvalbumin interneurons generate neuronal oscillatory activity in the gamma frequency range (30-80Hz) and gamma oscillations are closely linked with higher cognitive functions. Raloxifene, a selective estrogen receptor modulator, shows beneficial effects on human cognitive performance, and has few peripheral side effects unlike estradiol, but the biological mechanisms which underpin these benefits are not clear. This study aimed to investigate whether estradiol and raloxifene modulate hippocampal gamma-band oscillations during spatial memory performance. Prepubescent female mice were ovariectomized (OVX) and implanted with a subcutaneous pellet of either estradiol (E2), raloxifene or placebo. During adulthood, local field potentials were recorded from the dorsal hippocampus while mice were performing the Y-maze hippocampus-dependent spatial memory task. Ovariectomy caused deficits in spatial memory, accompanied by a significant reduction in hippocampal gamma oscillations, specifically during decision making. Estradiol as well as raloxifene rescued both behavioural and electrophysiological deficits. These data have significant implications for disorders of cognitive impairment where altered gamma oscillations are apparent, such as schizophrenia.
Collapse
|
29
|
Marques-Lopes J, Tesfaye E, Israilov S, Van Kempen TA, Wang G, Glass MJ, Pickel VM, Iadecola C, Waters EM, Milner TA. Redistribution of NMDA Receptors in Estrogen-Receptor-β-Containing Paraventricular Hypothalamic Neurons following Slow-Pressor Angiotensin II Hypertension in Female Mice with Accelerated Ovarian Failure. Neuroendocrinology 2017; 104:239-256. [PMID: 27078860 PMCID: PMC5381723 DOI: 10.1159/000446073] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/09/2016] [Indexed: 12/11/2022]
Abstract
Hypertension in male and aging female rodents is associated with glutamate-dependent plasticity in the hypothalamus, but existing models have failed to capture distinct transitional menopausal phases that could have a significant impact on the synaptic plasticity and emergent hypertension. In rodents, accelerated ovarian failure (AOF) induced by systemic injection of 4-vinylcyclohexane diepoxide mimics the estrogen fluctuations seen in human menopause including the perimenopause transition (peri-AOF) and postmenopause (post-AOF). Thus, we used the mouse AOF model to determine the impact of slow-pressor angiotensin II (AngII) administration on blood pressure and on the subcellular distribution of obligatory N-methyl-D-aspartate (NMDA) receptor GluN1 subunits in the paraventricular hypothalamic nucleus (PVN), a key estrogen-responsive cardiovascular regulatory area. Estrogen-sensitive neuronal profiles were identified in mice expressing enhanced green fluorescent protein under the promoter for estrogen receptor (ER) β, a major ER in the PVN. Slow-pressor AngII increased arterial blood pressure in mice at peri- and post-AOF time points. In control oil-injected (nonhypertensive) mice, AngII decreased the total number of GluN1 in ERβ-containing PVN dendrites. In contrast, AngII resulted in a reapportionment of GluN1 from the cytoplasm to the plasma membrane of ERβ-containing PVN dendrites in peri-AOF mice. Moreover, in post-AOF mice, AngII increased total GluN1, dendritic size and radical production in ERβ-containing neurons. These results indicate that unique patterns of hypothalamic glutamate receptor plasticity and dendritic structure accompany the elevated blood pressure in peri- and post-AOF time points. Our findings suggest the possibility that distinct neurobiological processes are associated with the increased blood pressure during perimenopausal and postmenopausal periods.
Collapse
Affiliation(s)
- Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, The Rockefeller University, New York, N.Y., USA
| | - Ephrath Tesfaye
- Feil Family Brain and Mind Research Institute, The Rockefeller University, New York, N.Y., USA
| | - Sigal Israilov
- Feil Family Brain and Mind Research Institute, The Rockefeller University, New York, N.Y., USA
| | - Tracey A. Van Kempen
- Feil Family Brain and Mind Research Institute, The Rockefeller University, New York, N.Y., USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, The Rockefeller University, New York, N.Y., USA
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, The Rockefeller University, New York, N.Y., USA
| | - Michael J. Glass
- Feil Family Brain and Mind Research Institute, The Rockefeller University, New York, N.Y., USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, The Rockefeller University, New York, N.Y., USA
| | - Virginia M. Pickel
- Feil Family Brain and Mind Research Institute, The Rockefeller University, New York, N.Y., USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, The Rockefeller University, New York, N.Y., USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, The Rockefeller University, New York, N.Y., USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, The Rockefeller University, New York, N.Y., USA
| | - Elizabeth M. Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, N.Y., USA
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, The Rockefeller University, New York, N.Y., USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, The Rockefeller University, New York, N.Y., USA
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, N.Y., USA
| |
Collapse
|
30
|
Nguyen TV, Ducharme S, Karama S. Effects of Sex Steroids in the Human Brain. Mol Neurobiol 2016; 54:7507-7519. [PMID: 27822715 DOI: 10.1007/s12035-016-0198-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 10/11/2016] [Indexed: 02/01/2023]
Abstract
Sex steroids are thought to play a critical developmental role in shaping both cortical and subcortical structures in the human brain. Periods of profound changes in sex steroids invariably coincide with the onset of sex differences in mental health vulnerability, highlighting the importance of sex steroids in determining sexual differentiation of the brain. Yet, most of the evidence for the central effects of sex steroids relies on non-human studies, as several challenges have limited our understanding of these effects in humans: the lack of systematic assessment of the human sex steroid metabolome, the different developmental trajectories of specific sex steroids, the impact of genetic variation and epigenetic changes, and the plethora of interactions between sex steroids, sex chromosomes, neurotransmitters, and other hormonal systems. Here we review how multimodal strategies may be employed to bridge the gap between the basic and clinical understanding of sex steroid-related changes in the human brain.
Collapse
Affiliation(s)
- Tuong-Vi Nguyen
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, QC, H3A 1A1, Canada.,Department of Obstetrics-Gynecology, McGill University Health Centre, McGill University, Montreal, QC, H3A 1A1, Canada
| | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, QC, H3A 1A1, Canada.,McConnell Brain Imaging Center, Montreal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Sherif Karama
- McConnell Brain Imaging Center, Montreal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada. .,Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, Montreal, QC, H4H 1R3, Canada.
| |
Collapse
|
31
|
Gopinath K, Maltbie E, Urushino N, Kempf D, Howell L. Ketamine-induced changes in connectivity of functional brain networks in awake female nonhuman primates: a translational functional imaging model. Psychopharmacology (Berl) 2016; 233:3673-3684. [PMID: 27530989 DOI: 10.1007/s00213-016-4401-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/28/2016] [Indexed: 12/13/2022]
Abstract
RATIONALE There is a significant interest in the NMDA-receptor antagonist ketamine due to its efficacy in treating depressive disorders and its induction of psychotic-like symptoms that make it a useful tool for modeling psychosis. Pharmacological MRI in awake nonhuman primates provides a highly translational model for studying the brain network dynamics involved in producing these drug effects. OBJECTIVE The present study evaluated ketamine-induced changes in functional connectivity (FC) in awake rhesus monkeys. The effects of ketamine after pretreatment with the antipsychotic drug risperidone were also examined. METHODS Functional MRI scans were conducted in four awake adult female rhesus monkeys during sub-anesthetic i.v. infusions of ketamine (0.345 mg/kg bolus followed by 0.256 mg kg-1 h-1 constant infusion) with and without risperidone pretreatment (0.06 mg/kg). A 10-min window of stable BOLD signal was used to compare FC between baseline and drug conditions. FC was assessed in specific regions of interest using seed-based cross-correlation analysis. RESULTS Ketamine infusion induced extensive changes in FC. In particular, FC to the dorsolateral prefrontal cortex (dlPFC) was increased in several cortical and subcortical regions. Pretreatment with risperidone largely attenuated ketamine-induced changes in FC. CONCLUSIONS The results are highly consistent with similar human imaging studies showing ketamine-induced changes in FC, as well as a significant attenuation of these changes when ketamine infusion is preceded by pretreatment with risperidone. The extensive increases shown in FC to the dlPFC are consistent with the idea that disinhibition of the dlPFC may be a key driver of the antidepressant and psychotomimetic effects of ketamine.
Collapse
Affiliation(s)
- Kaundinya Gopinath
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30329, USA.,Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd NE, Atlanta, GA, 30329, USA
| | - Eric Maltbie
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd NE, Atlanta, GA, 30329, USA
| | - Naoko Urushino
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd NE, Atlanta, GA, 30329, USA.,Dainippon Sumitomo Pharma, Co. Ltd., Osaka, Japan
| | - Doty Kempf
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd NE, Atlanta, GA, 30329, USA
| | - Leonard Howell
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd NE, Atlanta, GA, 30329, USA. .,Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, 30329, USA.
| |
Collapse
|
32
|
Al-Sweidi S, Morissette M, Di Paolo T. Estrogen receptors modulate striatal metabotropic receptor type 5 in intact and MPTP male mice model of Parkinson's disease. J Steroid Biochem Mol Biol 2016; 161:84-91. [PMID: 26873133 DOI: 10.1016/j.jsbmb.2016.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 12/23/2015] [Accepted: 02/07/2016] [Indexed: 01/17/2023]
Abstract
Glutamate is the most important brain excitatory neurotransmitter and glutamate overactivity is well documented in Parkinson's disease (PD). Metabotropic glutamate (mGlu) receptors are reported to interact with membrane estrogen receptors (ERs) and more specifically the mGlu5 receptor subtype. 17β-estradiol and mGlu5 antagonists have neuroprotective effects in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. We previously reported that ERα and ERβ are involved in neuroprotection following MPTP toxicity. The present study investigated the implication of ERs on the mGlu5 receptor adaptive response to MPTP toxicity in the brain of wild type (WT), ER knockout (ERKO)α and ERKOβ male mice. Autoradiography of [(3)H]ABP688 specific binding to striatal mGlu5 receptors showed a dorsal/ventral gradient similar for WT, ERKOα and ERKOβ mice with higher values ventrally. The lateral septum had highest [(3)H]ABP688 specific binding that remained unchanged in all experimental groups. ERKOα and ERKOβ mice had similarly lower striatal [(3)H]ABP688 specific binding than WT mice as measured also by Western blots. MPTP dose-dependently decreased striatal [(3)H]ABP688 specific binding in WT but not in ERKOα and ERKOβ mice; this correlated positively with striatal dopamine concentrations. A 17β-estradiol treatment for 10 days left unchanged striatal [(3)H]ABP688 specific binding of unlesioned mice of the three genotypes. 17β-estradiol treatment for 5 days before MPTP and for 5 days after partially prevented the mGlu5 receptor decrease only in WT MPTP mice and this was associated with higher BDNF striatal contents. These results thus show that in male mice ERs affect striatal mGlu5 receptor levels and their response to MPTP.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- Animals
- Brain-Derived Neurotrophic Factor/metabolism
- Corpus Striatum/metabolism
- Corpus Striatum/pathology
- Disease Models, Animal
- Estradiol/metabolism
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Estrogen Receptor beta/genetics
- Estrogen Receptor beta/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Parkinson Disease, Secondary/genetics
- Parkinson Disease, Secondary/metabolism
- Parkinson Disease, Secondary/pathology
- Receptor, Metabotropic Glutamate 5/analysis
- Receptor, Metabotropic Glutamate 5/metabolism
Collapse
Affiliation(s)
- S Al-Sweidi
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Quebec City, Quebec G1K 7P4, Canada
| | - M Morissette
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec G1V 4G2, Canada
| | - T Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec G1V 4G2, Canada; Faculty of Pharmacy, Laval University, Quebec City, Quebec G1K 7P4, Canada.
| |
Collapse
|
33
|
Khan MM. Neurocognitive, Neuroprotective, and Cardiometabolic Effects of Raloxifene: Potential for Improving Therapeutic Outcomes in Schizophrenia. CNS Drugs 2016; 30:589-601. [PMID: 27193386 DOI: 10.1007/s40263-016-0343-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Raloxifene is a selective estrogen receptor modulator that has been approved for treating osteoporosis and breast cancer in high-risk postmenopausal women. However, recent evidence suggests that raloxifene adjunct therapy improves cognition and reduces symptom severity in men and women with schizophrenia. In animal models, raloxifene increases forebrain neurogenesis and enhances working memory and synaptic plasticity. It may consequently repair the neuronal and synaptic connectivity that is disrupted in schizophrenia. It also reduces oxidative stress and neuroinflammation, which are potent etiological factors in the neuropathology of schizophrenia. Furthermore, in postmenopausal women, raloxifene reduces the risks for atherosclerosis, diabetes mellitus, and weight gain, which are serious adverse effects associated with long-term antipsychotic treatment in schizophrenia; therefore, it may improve the safety and efficacy of antipsychotic drugs. In this review, recent insights into the neurocognitive, neuroprotective, and cardiometabolic effects of raloxifene in relation to therapeutic outcomes in schizophrenia are discussed.
Collapse
Affiliation(s)
- Mohammad M Khan
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Zawia, Jamal Abdul Nassre Street, P.O. Box 16418, Az-Zawiyah, Libya.
| |
Collapse
|
34
|
Abstract
UNLABELLED A decline in estradiol (E2)-mediated cognitive benefits denotes a critical window for the therapeutic effects of E2, but the mechanism for closing of the critical window is unknown. We hypothesized that upregulating the expression of estrogen receptor α (ERα) or estrogen receptor β (ERβ) in the hippocampus of aged animals would restore the therapeutic potential of E2 treatments and rejuvenate E2-induced hippocampal plasticity. Female rats (15 months) were ovariectomized, and, 14 weeks later, adeno-associated viral vectors were used to express ERα, ERβ, or green fluorescent protein (GFP) in the CA1 region of the dorsal hippocampus. Animals were subsequently treated for 5 weeks with cyclic injections of 17β-estradiol-3-benzoate (EB, 10 μg) or oil vehicle. Spatial memory was examined 48 h after EB/oil treatment. EB treatment in the GFP (GFP + EB) and ERβ (ERβ + EB) groups failed to improve episodic spatial memory relative to oil-treated animals, indicating closing of the critical window. Expression of ERβ failed to improve cognition and was associated with a modest learning impairment. Cognitive benefits were specific to animals expressing ERα that received EB treatment (ERα + EB), such that memory was improved relative to ERα + oil and GFP + EB. Similarly, ERα + EB animals exhibited enhanced NMDAR-mediated synaptic transmission compared with the ERα + oil and GFP + EB groups. This is the first demonstration that the window for E2-mediated benefits on cognition and hippocampal E2 responsiveness can be reinstated by increased expression of ERα. SIGNIFICANCE STATEMENT Estradiol is neuroprotective, promotes synaptic plasticity in the hippocampus, and protects against cognitive decline associated with aging and neurodegenerative diseases. However, animal models and clinical studies indicate a critical window for the therapeutic treatment such that the beneficial effects are lost with advanced age and/or with extended hormone deprivation. We used gene therapy to upregulate expression of the estrogen receptors ERα and ERβ and demonstrate that the window for estradiol's beneficial effects on memory and hippocampal synaptic function can be reinstated by enhancing the expression of ERα. Our findings suggest that the activity of ERα controls the therapeutic window by regulating synaptic plasticity mechanisms involved in memory.
Collapse
|
35
|
Ketamine-induced brain activation in awake female nonhuman primates: a translational functional imaging model. Psychopharmacology (Berl) 2016; 233:961-72. [PMID: 26660447 PMCID: PMC4761287 DOI: 10.1007/s00213-015-4175-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/26/2015] [Indexed: 12/28/2022]
Abstract
RATIONALE There is significant interest in the NMDA receptor antagonist ketamine due to its efficacy in treating depressive disorders and its induction of psychotic-like symptoms that make it a useful tool for modeling psychosis. OBJECTIVE The present study extends the successful development of an apparatus and methodology to conduct pharmacological MRI studies in awake rhesus monkeys in order to evaluate the CNS effects of ketamine. METHODS Functional MRI scans were conducted in four awake adult female rhesus monkeys during sub-anesthetic intravenous (i.v.) infusions of ketamine (0.345 mg/kg bolus followed by 0.256 mg/kg/h constant infusion) with and without risperidone pretreatment (0.06 mg/kg). Statistical parametric maps of ketamine-induced blood oxygenation level-dependent (BOLD) activation were obtained with appropriate general linear regression models (GLMs) incorporating motion and hemodynamics of ketamine infusion. RESULTS Ketamine infusion induced and sustained robust BOLD activation in a number of cortical and subcortical regions, including the thalamus, cingulate gyrus, and supplementary motor area. Pretreatment with the antipsychotic drug risperidone markedly blunted ketamine-induced activation in many brain areas. CONCLUSIONS The results are remarkably similar to human imaging studies showing ketamine-induced BOLD activation in many of the same brain areas, and pretreatment with risperidone or another antipsychotic blunting the ketamine response to a similar extent. The strong concordance of the functional imaging data in humans with these results from nonhuman primates highlights the translational value of the model and provides an excellent avenue for future research examining the CNS effects of ketamine. This model may also be a useful tool for evaluating the efficacy of novel antipsychotic drugs.
Collapse
|
36
|
Morphological Differentiation Towards Neuronal Phenotype of SH-SY5Y Neuroblastoma Cells by Estradiol, Retinoic Acid and Cholesterol. Neurochem Res 2015; 41:731-47. [PMID: 26518675 PMCID: PMC4824837 DOI: 10.1007/s11064-015-1743-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 10/16/2015] [Accepted: 10/17/2015] [Indexed: 12/29/2022]
Abstract
Human SH-SY5Y neuroblastoma cells maintain their potential for differentiation and regression in culture conditions. The induction of differentiation could serve as a strategy to inhibit cell proliferation and tumor growth. Previous studies have shown that differentiation of SH-SY5Y cells can be induced by all-trans-retinoic-acid (RA) and cholesterol (CHOL). However, signaling pathways that lead to terminal differentiation of SH-SY5Y cells are still largely unknown. The goal of this study was to examine in the RA and CHOL treated SH-SY5Y cells the additive impacts of estradiol (E2) and brain-derived neurotrophic factor (BDNF) on cell morphology, cell population growth, synaptic vesicle recycling and presence of neurofilaments. The above features indicate a higher level of neuronal differentiation. Our data show that treatment for 10 days in vitro (DIV) with RA alone or when combined with E2 (RE) or CHOL (RC), but not when combined with BDNF (RB), significantly (p < 0.01) inhibited the cell population growth. Synaptic vesicle recycling, induced by high-K(+) depolarization, was significantly increased in all treatments where RA was included (RE, RC, RB, RCB), and when all agents were added together (RCBE). Specifically, our results show for the first time that E2 treatment can alone increase synaptic vesicle recycling in SH-SY5Y cells. This work contributes to the understanding of the ways to improve suppression of neuroblastoma cells' population growth by inducing maturation and differentiation.
Collapse
|
37
|
Gogos A, Sbisa AM, Sun J, Gibbons A, Udawela M, Dean B. A Role for Estrogen in Schizophrenia: Clinical and Preclinical Findings. Int J Endocrinol 2015; 2015:615356. [PMID: 26491441 PMCID: PMC4600562 DOI: 10.1155/2015/615356] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/21/2015] [Accepted: 08/23/2015] [Indexed: 12/30/2022] Open
Abstract
Gender differences in schizophrenia have been extensively researched and it is being increasingly accepted that gonadal steroids are strongly attributed to this phenomenon. Of the various hormones implicated, the estrogen hypothesis has been the most widely researched one and it postulates that estrogen exerts a protective effect by buffering females against the development and severity of the illness. In this review, we comprehensively analyse studies that have investigated the effects of estrogen, in particular 17β-estradiol, in clinical, animal, and molecular research with relevance to schizophrenia. Specifically, we discuss the current evidence on estrogen dysfunction in schizophrenia patients and review the clinical findings on the use of estradiol as an adjunctive treatment in schizophrenia patients. Preclinical research that has used animal models and molecular probes to investigate estradiol's underlying protective mechanisms is also substantially discussed, with particular focus on estradiol's impact on the major neurotransmitter systems implicated in schizophrenia, namely, the dopamine, serotonin, and glutamate systems.
Collapse
Affiliation(s)
- Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Alyssa M. Sbisa
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC 3086, Australia
| | - Jeehae Sun
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC 3086, Australia
| | - Andrew Gibbons
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Madhara Udawela
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Brian Dean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
38
|
Korol DL, Pisani SL. Estrogens and cognition: Friends or foes?: An evaluation of the opposing effects of estrogens on learning and memory. Horm Behav 2015; 74:105-15. [PMID: 26149525 PMCID: PMC4573330 DOI: 10.1016/j.yhbeh.2015.06.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/19/2015] [Accepted: 06/25/2015] [Indexed: 12/19/2022]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Estrogens are becoming well known for their robust enhancement on cognition particularly for learning and memory that relies upon functioning of the hippocampus and related neural systems. What is also emerging is that estrogen modulation of cognition is not uniform, at times enhancing yet at other times impairing learning. This review explores the bidirectional effects of estrogens on learning from a multiple memory systems view, focusing on the hippocampus and striatum, whereby modulation by estrogens sorts according to task attributes and neural systems engaged during cognition. We highlight our findings showing that the ability to solve hippocampus-sensitive tasks typically improves under relatively high estrogen status while the ability to solve striatum-sensitive tasks degrades with estrogen exposures. Though constrained by dose and timing of exposure, these opposing enhancements and impairments of cognition can be observed following treatments with different estrogenic compounds including the hormone estradiol, the isoflavone genistein found in soybeans, and agonists that are selective for specific estrogen receptors, suggesting that activation of a single receptor type is sufficient to produce the observed shifts in learning strategies. Using this multi-dimensional framework will allow us to extend our thinking of the relationship between estrogens and cognition to other brain regions and cognitive functions.
Collapse
Affiliation(s)
- Donna L Korol
- Department of Biology, Syracuse University, Department of Neuroscience and Physiology, SUNY-Upstate Medical University, Syracuse, NY 13244, USA.
| | - Samantha L Pisani
- Neuroscience Program and Medical Scholars Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
39
|
Ebrahimzadeh Bideskan AR, Lale Ataei M, Mansouri S, Hosseini M. The effects of tamoxifen and soy on dark neuron production in hippocampal formation after pentylenetetrazole-induced repeated seizures in rats. PATHOPHYSIOLOGY 2015; 22:125-35. [DOI: 10.1016/j.pathophys.2015.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 04/10/2015] [Accepted: 04/30/2015] [Indexed: 11/25/2022] Open
|
40
|
Sex-specific alterations in behavioral and cognitive functions in a “three hit” animal model of schizophrenia. Behav Brain Res 2015; 284:85-93. [DOI: 10.1016/j.bbr.2015.02.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 02/04/2015] [Accepted: 02/06/2015] [Indexed: 02/08/2023]
|
41
|
Quintana L, Harvey-Girard E, Lescano C, Macadar O, Lorenzo D. Sex-specific role of a glutamate receptor subtype in a pacemaker nucleus controlling electric behavior. ACTA ACUST UNITED AC 2014; 108:155-66. [PMID: 24794754 DOI: 10.1016/j.jphysparis.2014.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/12/2014] [Accepted: 04/22/2014] [Indexed: 02/03/2023]
Abstract
Electric communication signals, produced by South American electric fish, vary across sexes and species and present an ideal opportunity to examine the bases of signal diversity, and in particular, the mechanisms underlying sexually dimorphic behavior. Gymnotiforms produce electric organ discharges (EOD) controlled by a hindbrain pacemaker nucleus (PN). Background studies have identified the general cellular mechanisms that underlie the production of communication signals, EOD chirps and interruptions, typically displayed in courtship and agonistic contexts. Brachyhypopomus gauderio emit sexually dimorphic signals, and recent studies have shown that the PN acquires the capability of generating chirps seasonally, only in breeding males, by modifying its glutamatergic system. We hypothesized that sexual dimorphism was caused by sexual differences in the roles of glutamate receptors. To test this hypothesis, we analyzed NMDA and AMPA mediated responses in PN slice preparations by field potential recordings, and quantified one AMPA subunit mRNA, in the PNs of males and females during the breeding season. In situ hybridization of GluR2B showed no sexual differences in quantities between the male and female PN. Functional responses of the PN to glutamate and AMPA, on the other hand, showed a clear cut sexual dimorphism. In breeding males, but not females, the PN responded to glutamate and AMPA with bursting activity, with a temporal pattern that resembled the pattern of EOD chirps. In this study, we have been successful in identifying cellular mechanisms of sexual dimorphic communication signals. The involvement of AMPA receptors in PN activity is part of the tightly regulated changes that account for the increase in signal diversity during breeding in this species, necessary for a successful reproduction.
Collapse
Affiliation(s)
- Laura Quintana
- Unidad Bases Neurales de la Conducta, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay.
| | - Erik Harvey-Girard
- Department of Cell and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Carolina Lescano
- Unidad Bases Neurales de la Conducta, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Omar Macadar
- Unidad Bases Neurales de la Conducta, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Daniel Lorenzo
- Unidad Bases Neurales de la Conducta, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
42
|
Bian C, Zhu H, Zhao Y, Cai W, Zhang J. Intriguing roles of hippocampus-synthesized 17β-estradiol in the modulation of hippocampal synaptic plasticity. J Mol Neurosci 2014; 54:271-81. [PMID: 24729128 DOI: 10.1007/s12031-014-0285-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/06/2014] [Indexed: 10/25/2022]
Abstract
Accumulated studies have shown that 17β-estradiol (E2) can be de novo synthesized in the hippocampus, and its role in the regulation of hippocampal synaptic plasticity, which is the basis of learning and memory, has long been exploring. Steroidogenic enzymes (e.g., aromatase) that are essential to the hippocampus-synthesized synthesis of E2 have been detected in the hippocampus. Inhibition of E2 synthesis by aromatase inhibitors significantly reduces the density of hippocampal spine synapses, levels of some synaptic proteins such as spinopholin and synaptophysin. Moreover, the electrophysiological properties of hippocampal neurons are also changed in response to this inhibition. The influences of gonadal and hippocampal E2 on synaptic plasticity may exist some differences, since some reports showed that gonadal (or circulating) estrogens have no obvious effects in the modulation of hippocampal synaptic proteins as evidenced in some ovariectomized animals and postmenopausal women who suffered from Alzheimer's disease (AD). These evidences leads to a hypothesis that hippocampal E2 may play a more important role in modulation of synaptic plasticity than gonadal E2. The signaling pathways, whereby hippocampal E2 modulates synaptic plasticity, insist of classical chronic genomic pathway and rapid nongenomic pathway, which mediated by nonnuclear estrogen receptor (GPER) and/or nuclear or nonnuclear estrogen receptors, which require coactivators for their transcription activity. Among which steroid receptor coactivator-1 (SRC-1) is the predominant coactivator p160 family members in the brain. Several clues have shown that SRC-1 is expressed in hippocampus and is highly correlated with some key synaptic proteins developmentally or after orchidectomy but not ovariectomy, indicating SRC-1 may be regulated by hippocampus-synthesized E2 and profoundly involved in the mediation of hippocampal E2 regulation of hippocampal synaptic plasticity. Further studies about the exact roles of hippocampus-synthesized E2 and therefore SRC-1 are urgently needed in order to facilitate our understanding of hippocampal E2, which will be very important to the development of novel strategies of estrogen replacement therapy against neurodegenerative deficits such as Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Chen Bian
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Gaotanyan 30, Chongqing, 400038, China
| | | | | | | | | |
Collapse
|
43
|
St John JA, Henderson VW, Hodis HN, Kono N, McCleary CA, Franke AA, Mack WJ. Associations between urine excretion of isoflavonoids and cognition in postmenopausal women in the Women's Isoflavone Soy Health clinical trial. J Am Geriatr Soc 2014; 62:629-35. [PMID: 24617349 DOI: 10.1111/jgs.12752] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OBJECTIVES To determine effect of change in urine excretion of isoflavonoids on cognitive change. DESIGN Post hoc analysis of isoflavonoid exposure (mean 2.7 years) during the randomized, placebo-controlled, double-blind Women's Isoflavone Soy Health trial. SETTING General community. PARTICIPANTS Healthy postmenopausal women (N = 350). INTERVENTION Twenty-five grams of isoflavone-rich soy protein (91 mg of aglycone weight isoflavones: 52 mg genistein, 36 mg daidzein, 3 mg glycitein) or milk protein-matched placebo provided daily. MEASUREMENTS Overnight urine excretion, fasting plasma levels of isoflavonoids, and cognitive function measured at baseline and endpoint. RESULTS Three hundred women (age: mean 61, range 45-92) completed both cognitive assessments and did not use hormone replacement therapy during the trial. Mean on-trial change from baseline in urine excretion of isoflavonoids was not significantly associated with change in a composite score of global cognition (P = .39). Secondary analyses indicated that change in urine excretion of isoflavonoids was inversely associated with change in a factor score representing general intelligence (P = .02) but not with factor scores representing verbal or visual episodic memory. Mean differences in this general intelligence factor score between women in the lowest and highest quartiles of isoflavonoid change were equivalent to an approximate 4.4-year age-associated decline. Analyses based on plasma isoflavonoid levels yielded similar but attenuated results. CONCLUSION In healthy postmenopausal women, long-term changes in isoflavonoids are not associated with global cognition, supporting clinical trial results, although greater isoflavonoid exposure from dietary supplements is associated with decrements in general intelligence but not memory; this finding requires confirmation in future studies.
Collapse
Affiliation(s)
- Jan A St John
- Atherosclerosis Research Unit, University of Southern California, Los Angeles, California; Department of Preventive Medicine, University of Southern California, Los Angeles, California
| | | | | | | | | | | | | |
Collapse
|
44
|
Newhouse P, Albert K, Astur R, Johnson J, Naylor M, Dumas J. Tamoxifen improves cholinergically modulated cognitive performance in postmenopausal women. Neuropsychopharmacology 2013; 38:2632-43. [PMID: 23867982 PMCID: PMC3828534 DOI: 10.1038/npp.2013.172] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 06/25/2013] [Accepted: 07/02/2013] [Indexed: 12/15/2022]
Abstract
Tamoxifen (TMX) is a selective estrogen receptor modulator that is used as an estrogen receptor antagonist for the treatment and prevention of breast cancer. Whether TMX has antagonist activities in the human brain is less clear and its effects on cognitive function have not been experimentally explored. This study examined how TMX affected cognitive performance in older women using a model of anticholinergic drug-induced cognitive dysfunction. Twenty-one postmenopausal women were administered 20 mg of oral TMX or placebo for 3 months. Participants then took part in five drug challenges using the anticholinergic antinicotinic agent mecamylamine (MECA) and antimuscarinic agent scopolamine (SCOP) and were tested on a comprehensive battery including tasks of attention and psychomotor function, verbal episodic memory, and spatial navigation. After a 3-month placebo washout, participants were then crossed over to the alternate treatment and repeated the drug challenges after 3 months. Compared with placebo treatment, TMX significantly attenuated the impairment from cholinergic blockade on tasks of verbal episodic memory and spatial navigation, but effects on attentional/psychomotor tasks were more variable. Analysis by APOE genotype showed that APO ɛ4+ women showed a greater beneficial effect of TMX on reversing the cholinergic impairment than APO ɛ4- women on most tasks. This study provides evidence that TMX may act as an estrogen-like agonist to enhance cholinergic system activity and hippocampally mediated learning.
Collapse
Affiliation(s)
- Paul Newhouse
- Vanderbilt Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA,Clinical Neuroscience Research Unit, Department of Psychiatry, University of Vermont College of Medicine, Burlington, VT, USA,Vanderbilt Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University School of Medicine, 1601 23rd Avenue, Nashville, TN 37212, USA, Tel: +1 615 936 0928, Fax: +1 615 875 0686, E-mail:
| | - Kimberly Albert
- Vanderbilt Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert Astur
- Department of Psychology, University of Connecticut, Storrs, CT, USA
| | - Julia Johnson
- Department of Obstetrics and Gynecology, University of Massachusetts School of Medicine, Worcester, MA, USA
| | - Magdalena Naylor
- Clinical Neuroscience Research Unit, Department of Psychiatry, University of Vermont College of Medicine, Burlington, VT, USA
| | - Julie Dumas
- Clinical Neuroscience Research Unit, Department of Psychiatry, University of Vermont College of Medicine, Burlington, VT, USA
| |
Collapse
|
45
|
Banerjee SB, Liu RC. Storing maternal memories: hypothesizing an interaction of experience and estrogen on sensory cortical plasticity to learn infant cues. Front Neuroendocrinol 2013; 34:300-14. [PMID: 23916405 PMCID: PMC3788048 DOI: 10.1016/j.yfrne.2013.07.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/18/2013] [Accepted: 07/22/2013] [Indexed: 11/15/2022]
Abstract
Much of the literature on maternal behavior has focused on the role of infant experience and hormones in a canonical subcortical circuit for maternal motivation and maternal memory. Although early studies demonstrated that the cerebral cortex also plays a significant role in maternal behaviors, little has been done to explore what that role may be. Recent work though has provided evidence that the cortex, particularly sensory cortices, contains correlates of sensory memories of infant cues, consistent with classical studies of experience-dependent sensory cortical plasticity in non-maternal paradigms. By reviewing the literature from both the maternal behavior and sensory cortical plasticity fields, focusing on the auditory modality, we hypothesize that maternal hormones (predominantly estrogen) may act to prime auditory cortical neurons for a longer-lasting neural trace of infant vocal cues, thereby facilitating recognition and discrimination. This couldthen more efficiently activate the subcortical circuit to elicit and sustain maternal behavior.
Collapse
Affiliation(s)
- Sunayana B. Banerjee
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322
| | - Robert C. Liu
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322
| |
Collapse
|
46
|
Mott NN, Pak TR. Estrogen signaling and the aging brain: context-dependent considerations for postmenopausal hormone therapy. ISRN ENDOCRINOLOGY 2013; 2013:814690. [PMID: 23936665 PMCID: PMC3725729 DOI: 10.1155/2013/814690] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 05/21/2013] [Indexed: 02/08/2023]
Abstract
Recent clinical studies have spurred rigorous debate about the benefits of hormone therapy (HT) for postmenopausal women. Controversy first emerged based on a sharp increase in the risk of cardiovascular disease in participants of the Women's Health Initiative (WHI) studies, suggesting that decades of empirical research in animal models was not necessarily applicable to humans. However, a reexamination of the data from the WHI studies suggests that the timing of HT might be a critical factor and that advanced age and/or length of estrogen deprivation might alter the body's ability to respond to estrogens. Dichotomous estrogenic effects are mediated primarily by the actions of two high-affinity estrogen receptors alpha and beta (ER α & ER β ). The expression of the ERs can be overlapping or distinct, dependent upon brain region, sex, age, and exposure to hormone, and, during the time of menopause, there may be changes in receptor expression profiles, post-translational modifications, and protein:protein interactions that could lead to a completely different environment for E2 to exert its effects. In this review, factors affecting estrogen-signaling processes will be discussed with particular attention paid to the expression and transcriptional actions of ER β in brain regions that regulate cognition and affect.
Collapse
Affiliation(s)
- Natasha N. Mott
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, 2160 S First Avenue, Maywood, IL 60153, USA
| | - Toni R. Pak
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, 2160 S First Avenue, Maywood, IL 60153, USA
| |
Collapse
|
47
|
Zhang XH, Jia N, Zhao XY, Tang GK, Guan LX, Wang D, Sun HL, Li H, Zhu ZL. Involvement of pGluR1, EAAT2 and EAAT3 in offspring depression induced by prenatal stress. Neuroscience 2013; 250:333-41. [PMID: 23694703 DOI: 10.1016/j.neuroscience.2013.04.031] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 03/27/2013] [Accepted: 04/16/2013] [Indexed: 12/18/2022]
Abstract
It is widely known that prenatal stress (PS) exposure causes depression-like behaviour to offspring, as well as maladaptive responses including neurobiological and physiological changes. However, the underlying mechanism of PS induced juvenile-onset depression remains largely unravelled. The inadequacies of monoamine deficiency hypothesis, the emerging evidence of altered glutamate neurotransmission in mood disorders, as well as our previous studies inspired us to assess the potential role of glutamatergic system in the pathogenesis of juvenile depression. In this research, we examined the expression of phosphorylated GluR1 subunit of ionotropic receptor alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR), the Na+-dependent glutamate transporters excitatory amino acid transporter 2 (EAAT2) and EAAT3 in the hippocampus, striatum and frontal cortex of 1-month-old rat offspring after mid and late PS exposure. Prenatally stressed offspring rats showed significantly prolonged duration of immobility and shortened immobility latency in tail suspension test. We also detected that PS significantly altered the expression of glutamate receptor and glutamate transporters of these depressed rats. In brief, the changes of phosphorylated GluR1 subunit of AMPAR protein level in the hippocampus and frontal cortex, as well as markedly decreased EAAT2 mRNA expression in the hippocampus, striatum and frontal cortex and EAAT3 mRNA expression in the hippocampus of stressed rats were both observed. These results underpinned that glutamate receptors and glutamate transporters might be involved in the progress of depression-like behaviour in juvenile rat offspring induced by PS.
Collapse
Affiliation(s)
- X H Zhang
- Department of Pharmacology, College of Medicine, Xi'an Jiaotong University, 86-710061 Shaanxi, China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Demonstration of estrogen receptor α protein in glutamatergic (vesicular glutamate transporter 2 immunoreactive) neurons of the female rat hypothalamus and amygdala using double-label immunocytochemistry. Exp Brain Res 2013; 226:595-602. [DOI: 10.1007/s00221-013-3474-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 02/28/2013] [Indexed: 11/25/2022]
|
49
|
Al-Sweidi S, Morissette M, Di Paolo T. Effect of oestrogen receptors on brain NMDA receptors of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice. J Neuroendocrinol 2012; 24:1375-85. [PMID: 22672467 DOI: 10.1111/j.1365-2826.2012.02349.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Parkinson's disease (PD) is characterised by the loss of nigrostriatal dopamine (DA) neurones and glutamate overactivity. There is substantial evidence to suggest that oestrogens prevent or delay the disease. 17β-oestradiol has neuroprotective effects in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD and modulates brain NMDA receptors. In MPTP-lesioned mice, oestrogen receptor (ER)α and ERβ are important in 17β-oestradiol-induced neuroprotection. To evaluate the role of ERs in the response of NMDA receptors to lesion, we compared wild-type (WT) with ER knockout (KO) C57Bl/6 male mice that received 7, 9 or 11 mg/kg of MPTP. These mice were also treated with MPTP (9 mg/kg) and 17β-oestradiol. [(3) H]Ro 25-6981 specific binding autoradiography was used to label NMDA receptors containing NR2B subunits. In the frontal and cingulate cortex and striatum, vehicle-treated WT mice had higher [(3) H]Ro 25-6981 specific binding compared to ERKO mice. Cortical [(3) H]Ro 25-6981 specific binding decreased with increasing doses of MPTP in WT and ERKOα but not ERKOβ mice, whereas a dose-related decrease was only observed in the striatum of WT mice remaining low in ERKOα and ERKOβ mice. No effect of 17β-oestradiol treatment in intact or MPTP-lesioned mice of all three genotypes was observed in the cortex, whereas it increased striatal specific binding of intact ERKOβ and MPTP-lesioned WT mice. Striatal [(3) H]Ro 25-6981 specific binding positively correlated with striatal DA concentrations only in WT mice. MPTP and 17β-oestradiol treatments had more limited effects in the hippocampus. Only in the CA3 and dentate gyrus did vehicle and 17β-oestradiol-treated ERKOα mice have higher [(3) H]Ro 25-6981 specific binding than WT and ERKOβ mice, whereas MPTP decreased this specific binding only in the CA1, CA2 and CA3 of ERKOα mice. Hence, brain NMDA receptors were affected by the deletion of ERs, which affect the response to MPTP and 17β-oestradiol treatments with brain region specificity.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- Animals
- Brain/drug effects
- Brain/metabolism
- Brain/pathology
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/metabolism
- CA1 Region, Hippocampal/pathology
- Dose-Response Relationship, Drug
- Estradiol/blood
- Estradiol/pharmacology
- Gene Expression Regulation/drug effects
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Parkinsonian Disorders/chemically induced
- Parkinsonian Disorders/genetics
- Parkinsonian Disorders/metabolism
- Parkinsonian Disorders/pathology
- Phenols/pharmacology
- Piperidines/pharmacology
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/physiology
- Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
Collapse
Affiliation(s)
- S Al-Sweidi
- Endocrinology and Genomics Research Axis of the CHUQ, CHUL, Quebec City, Quebec, Canada
| | | | | |
Collapse
|
50
|
Ooishi Y, Kawato S, Hojo Y, Hatanaka Y, Higo S, Murakami G, Komatsuzaki Y, Ogiue-Ikeda M, Kimoto T, Mukai H. Modulation of synaptic plasticity in the hippocampus by hippocampus-derived estrogen and androgen. J Steroid Biochem Mol Biol 2012; 131:37-51. [PMID: 22075082 DOI: 10.1016/j.jsbmb.2011.10.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/27/2011] [Accepted: 10/12/2011] [Indexed: 12/29/2022]
Abstract
The hippocampus synthesizes estrogen and androgen in addition to the circulating sex steroids. Synaptic modulation by hippocampus-derived estrogen or androgen is essential to maintain healthy memory processes. Rapid actions (1-2h) of 17β-estradiol (17β-E2) occur via synapse-localized receptors (ERα or ERβ), while slow genomic E2 actions (6-48h) occur via classical nuclear receptors (ERα or ERβ). The long-term potentiation (LTP), induced by strong tetanus or theta-burst stimulation, is not further enhanced by E2 perfusion in adult rats. Interestingly, E2 perfusion can rescue corticosterone (stress hormone)-induced suppression of LTP. The long-term depression is modulated rapidly by E2 perfusion. Elevation of the E2 concentration changes rapidly the density and head structure of spines in neurons. ERα, but not ERβ, drives this enhancement of spinogenesis. Kinase networks are involved downstream of ERα. Testosterone (T) or dihydrotestosterone (DHT) also rapidly modulates spinogenesis. Newly developed Spiso-3D mathematical analysis is used to distinguish these complex effects by sex steroids and kinases. It has been doubted that the level of hippocampus-derived estrogen and androgen may not be high enough to modulate synaptic plasticity. Determination of the accurate concentration of E2, T or DHT in the hippocampus is enabled by mass-spectrometric analysis in combination with new steroid-derivatization methods. The E2 level in the hippocampus is approximately 8nM for the male and 0.5-2nM for the female, which is much higher than that in circulation. The level of T and DHT is also higher than that in circulation. Taken together, hippocampus-derived E2, T, and DHT play a major role in modulation of synaptic plasticity.
Collapse
Affiliation(s)
- Yuuki Ooishi
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|