1
|
Ganesan K, Ghorbanpour S, Kendall W, Broome ST, Gladding JM, Dhungana A, Abiero AR, Mahmoudi M, Castorina A, Kendig MD, Becchi S, Valova V, Cole L, Bradfield LA. Hippocampal neuroinflammation induced by lipopolysaccharide causes sex-specific disruptions in action selection, food approach memories, and neuronal activation. Brain Behav Immun 2025; 124:9-27. [PMID: 39547520 DOI: 10.1016/j.bbi.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/24/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024] Open
Abstract
Hippocampal neuroinflammation is present in multiple diseases and disorders that impact motivated behaviour in a sex-specific manner, but whether neuroinflammation alone is sufficient to disrupt this behaviour is unknown. We investigated this question here using mice. First, the application of an endotoxin to primary cultures containing only hippocampal neurons did not affect their activation. However, when the same endotoxin was applied to mixed neuronal/glial cultures it did increase neuronal activation, providing initial indications of how it might be able to effect behavioural change. We next showed neuroinflammatory effects on behaviour directly, demonstrating that intra-hippocampal administration of the same endotoxin increased locomotor activity and accelerated goal-directed learning in both male and female mice. In contrast, lipopolysaccharide-induced hippocampal neuroinflammation caused sex-specific disruptions to the acquisition of instrumental actions and to Pavlovian food-approach memories. Finally, we showed that LPS-induced hippocampal neuroinflammation had a sexually dimorphic effect on neuronal activation: increasing it in females and decreasing it in males.
Collapse
Affiliation(s)
- Kiruthika Ganesan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia; School of Psychology, Faculty of Science, University of Sydney, New South Wales 2006, Australia
| | - Sahar Ghorbanpour
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, BOKU University, Vienna, Austria
| | - William Kendall
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Sarah Thomas Broome
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France
| | - Joanne M Gladding
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Amolika Dhungana
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia
| | - Arvie Rodriguez Abiero
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia; School of Psychology, Faculty of Science, University of Sydney, New South Wales 2006, Australia
| | - Maedeh Mahmoudi
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Alessandro Castorina
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Michael D Kendig
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Serena Becchi
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia; Teva Pharmaceuticals, Sydney, New South Wales 2113, Australia
| | - Veronika Valova
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, New South Wales 2050, Australia
| | - Louise Cole
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Laura A Bradfield
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia.
| |
Collapse
|
2
|
Maroteaux MJ, Noccioli CT, Daniel JM, Schrader LA. Rapid and local neuroestrogen synthesis supports long-term potentiation of hippocampal Schaffer collaterals-cornu ammonis 1 synapse in ovariectomized mice. J Neuroendocrinol 2024; 36:e13450. [PMID: 39351868 DOI: 10.1111/jne.13450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/31/2024] [Accepted: 09/04/2024] [Indexed: 10/03/2024]
Abstract
In aging women, cognitive decline and increased risk of dementia have been associated with the cessation of ovarian hormones production at menopause. In the brain, presence of the key enzyme aromatase required for the synthesis of 17-β-estradiol (E2) allows for local production of E2 in absence of functional ovaries. Understanding how aromatase activity is regulated could help alleviate the cognitive symptoms. In female rodents, genetic or pharmacological reduction of aromatase activity over extended periods of time impair memory formation, decreases spine density, and hinders long-term potentiation (LTP) in the hippocampus. Conversely, increased excitatory neurotransmission resulting in rapid N-methyl-d-aspartic acid (NMDA) receptor activation rapidly promotes neuroestrogen synthesis. This rapid modulation of aromatase activity led us to address the hypothesis that acute neuroestrogens synthesis is necessary for LTP at the Schaffer collateral-cornu ammonis 1 (CA1) synapse in absence of circulating ovarian estrogens. To test this hypothesis, we did electrophysiological recordings of field excitatory postsynaptic potential (fEPSPs) in hippocampal slices obtained from ovariectomized mice. To assess the impact of neuroestrogens synthesis on LTP, we applied the specific aromatase inhibitor, letrozole, before the induction of LTP with a theta burst stimulation protocol. We found that blocking aromatase activity prevented LTP. Interestingly, exogenous E2 application, while blocking aromatase activity, was not sufficient to recover LTP in our model. Our results indicate the critical importance of rapid, activity-dependent local neuroestrogens synthesis, independent of circulating hormones for hippocampal synaptic plasticity in female rodents.
Collapse
Affiliation(s)
- Matthieu J Maroteaux
- Department of Psychology, Tulane University, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Claire T Noccioli
- Department of Psychology, Tulane University, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Jill M Daniel
- Department of Psychology, Tulane University, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Laura A Schrader
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
| |
Collapse
|
3
|
Bauer EP. Sex differences in fear responses: Neural circuits. Neuropharmacology 2023; 222:109298. [PMID: 36328063 PMCID: PMC11267399 DOI: 10.1016/j.neuropharm.2022.109298] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/26/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Women have increased vulnerability to PTSD and anxiety disorders compared to men. Understanding the neurobiological underpinnings of these disorders is critical for identifying risk factors and developing appropriate sex-specific interventions. Despite the clear clinical relevance of an examination of sex differences in fear responses, the vast majority of pre-clinical research on fear learning and memory formation has exclusively used male animals. This review highlights sex differences in context and cued fear conditioning, fear extinction and fear generalization with a focus on the neural circuits underlying these behaviors in rodents. There are mixed reports of behavioral sex differences in context and cued fear conditioning paradigms, which can depend upon the behavioral indices of fear. However, there is greater evidence of differential activation of the hippocampus, amygdalar nuclei and the prefrontal cortical regions in male and female rodents during context and cued fear conditioning. The bed nucleus of the stria terminalis (BNST), a sexually dimorphic structure, is of particular interest as it differentially contributes to fear responses in males and females. In addition, while the influence of the estrous cycle on different phases of fear conditioning is delineated, the clearest modulatory effect of estrogen is on fear extinction processes. Examining the variability in neural responses and behavior in both sexes should increase our understanding of how that variability contributes to the neurobiology of affective disorders. This article is part of the Special Issue on 'Fear, anxiety and PTSD'.
Collapse
Affiliation(s)
- Elizabeth P Bauer
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY, 10027, United States.
| |
Collapse
|
4
|
Saleki K, Banazadeh M, Saghazadeh A, Rezaei N. Aging, testosterone, and neuroplasticity: friend or foe? Rev Neurosci 2022; 34:247-273. [PMID: 36017670 DOI: 10.1515/revneuro-2022-0033] [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: 04/01/2022] [Accepted: 07/03/2022] [Indexed: 11/15/2022]
Abstract
Neuroplasticity or neural plasticity implicates the adaptive potential of the brain in response to extrinsic and intrinsic stimuli. The concept has been utilized in different contexts such as injury and neurological disease. Neuroplasticity mechanisms have been classified into neuroregenerative and function-restoring processes. In the context of injury, neuroplasticity has been defined in three post-injury epochs. Testosterone plays a key yet double-edged role in the regulation of several neuroplasticity alterations. Research has shown that testosterone levels are affected by numerous factors such as age, stress, surgical procedures on gonads, and pharmacological treatments. There is an ongoing debate for testosterone replacement therapy (TRT) in aging men; however, TRT is more useful in young individuals with testosterone deficit and more specific subgroups with cognitive dysfunction. Therefore, it is important to pay early attention to testosterone profile and precisely uncover its harms and benefits. In the present review, we discuss the influence of environmental factors, aging, and gender on testosterone-associated alterations in neuroplasticity, as well as the two-sided actions of testosterone in the nervous system. Finally, we provide practical insights for further study of pharmacological treatments for hormonal disorders focusing on restoring neuroplasticity.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,USERN Office, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| | - Mohammad Banazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, 76169 13555 Kerman, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 14176 13151 Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| |
Collapse
|
5
|
Kuwahara N, Nicholson K, Isaacs L, MacLusky NJ. Androgen Effects on Neural Plasticity. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:216-230. [PMID: 35024693 PMCID: PMC8744448 DOI: 10.1089/andro.2021.0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Androgens are synthesized in the brain, gonads, and adrenal glands, in both sexes, exerting physiologically important effects on the structure and function of the central nervous system. These effects may contribute to the incidence and progression of neurological disorders such as autism spectrum disorder, schizophrenia, and Alzheimer's disease, which occur at different rates in males and females. This review briefly summarizes the current state of knowledge with respect to the neuroplastic effects of androgens, with particular emphasis on the hippocampus, which has been the focus of much of the research in this field.
Collapse
Affiliation(s)
- Nariko Kuwahara
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kate Nicholson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Lauren Isaacs
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Neil J. MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
6
|
Brann DW, Lu Y, Wang J, Sareddy GR, Pratap UP, Zhang Q, Tekmal RR, Vadlamudi RK. Neuron-Derived Estrogen-A Key Neuromodulator in Synaptic Function and Memory. Int J Mol Sci 2021; 22:ijms222413242. [PMID: 34948039 PMCID: PMC8706511 DOI: 10.3390/ijms222413242] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 01/31/2023] Open
Abstract
In addition to being a steroid hormone, 17β-estradiol (E2) is also a neurosteroid produced in neurons in various regions of the brain of many species, including humans. Neuron-derived E2 (NDE2) is synthesized from androgen precursors via the action of the biosynthetic enzyme aromatase, which is located at synapses and in presynaptic terminals in neurons in both the male and female brain. In this review, we discuss evidence supporting a key role for NDE2 as a neuromodulator that regulates synaptic plasticity and memory. Evidence supporting an important neuromodulatory role of NDE2 in the brain has come from studies using aromatase inhibitors, aromatase overexpression in neurons, global aromatase knockout mice, and the recent development of conditional forebrain neuron-specific knockout mice. Collectively, these studies demonstrate a key role of NDE2 in the regulation of synapse and spine density, efficacy of excitatory synaptic transmission and long-term potentiation, and regulation of hippocampal-dependent recognition memory, spatial reference memory, and contextual fear memory. NDE2 is suggested to achieve these effects through estrogen receptor-mediated regulation of rapid kinase signaling and CREB-BDNF signaling pathways, which regulate actin remodeling, as well as transcription, translation, and transport of synaptic proteins critical for synaptic plasticity and function.
Collapse
Affiliation(s)
- Darrell W. Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Correspondence:
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Gangadhara R. Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (G.R.S.); (U.P.P.); (R.R.T.); (R.K.V.)
| | - Uday P. Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (G.R.S.); (U.P.P.); (R.R.T.); (R.K.V.)
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health, Shreveport, LA 71103, USA;
| | - Rajeshwar R. Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (G.R.S.); (U.P.P.); (R.R.T.); (R.K.V.)
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (G.R.S.); (U.P.P.); (R.R.T.); (R.K.V.)
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| |
Collapse
|
7
|
Brann DW, Lu Y, Wang J, Zhang Q, Thakkar R, Sareddy GR, Pratap UP, Tekmal RR, Vadlamudi RK. Brain-derived estrogen and neural function. Neurosci Biobehav Rev 2021; 132:793-817. [PMID: 34823913 PMCID: PMC8816863 DOI: 10.1016/j.neubiorev.2021.11.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/26/2021] [Accepted: 11/12/2021] [Indexed: 01/02/2023]
Abstract
Although classically known as an endocrine signal produced by the ovary, 17β-estradiol (E2) is also a neurosteroid produced in neurons and astrocytes in the brain of many different species. In this review, we provide a comprehensive overview of the localization, regulation, sex differences, and physiological/pathological roles of brain-derived E2 (BDE2). Much of what we know regarding the functional roles of BDE2 has come from studies using specific inhibitors of the E2 synthesis enzyme, aromatase, as well as the recent development of conditional forebrain neuron-specific and astrocyte-specific aromatase knockout mouse models. The evidence from these studies support a critical role for neuron-derived E2 (NDE2) in the regulation of synaptic plasticity, memory, socio-sexual behavior, sexual differentiation, reproduction, injury-induced reactive gliosis, and neuroprotection. Furthermore, we review evidence that astrocyte-derived E2 (ADE2) is induced following brain injury/ischemia, and plays a key role in reactive gliosis, neuroprotection, and cognitive preservation. Finally, we conclude by discussing the key controversies and challenges in this area, as well as potential future directions for the field.
Collapse
Affiliation(s)
- Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Roshni Thakkar
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Gangadhara R Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Uday P Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Rajeshwar R Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA; Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
| |
Collapse
|
8
|
Soutar CN, Grenier P, Patel A, Kabitsis PP, Olmstead MC, Bailey CDC, Dringenberg HC. Brain-Generated 17β-Estradiol Modulates Long-Term Synaptic Plasticity in the Primary Auditory Cortex of Adult Male Rats. Cereb Cortex 2021; 32:2140-2155. [PMID: 34628498 DOI: 10.1093/cercor/bhab345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Neuron-derived 17β-estradiol (E2) alters synaptic transmission and plasticity in brain regions with endocrine and non-endocrine functions. Investigations into a modulatory role of E2 in synaptic activity and plasticity have mainly focused on the rodent hippocampal formation. In songbirds, E2 is synthesized by auditory forebrain neurons and promotes auditory signal processing and memory for salient acoustic stimuli; however, the modulatory effects of E2 on memory-related synaptic plasticity mechanisms have not been directly examined in the auditory forebrain. We investigated the effects of bidirectional E2 manipulations on synaptic transmission and long-term potentiation (LTP) in the rat primary auditory cortex (A1). Immunohistochemistry revealed widespread neuronal expression of the E2 biosynthetic enzyme aromatase in multiple regions of the rat sensory and association neocortex, including A1. In A1, E2 application reduced the threshold for in vivo LTP induction at layer IV synapses, whereas pharmacological suppression of E2 production by aromatase inhibition abolished LTP induction at layer II/III synapses. In acute A1 slices, glutamate and γ-aminobutyric acid (GABA) receptor-mediated currents were sensitive to E2 manipulations in a layer-specific manner. These findings demonstrate that locally synthesized E2 modulates synaptic transmission and plasticity in A1 and suggest potential mechanisms by which E2 contributes to auditory signal processing and memory.
Collapse
Affiliation(s)
- Chloe N Soutar
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Patrick Grenier
- Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Ashutosh Patel
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Pauline P Kabitsis
- Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Mary C Olmstead
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada.,Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Craig D C Bailey
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Hans C Dringenberg
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada.,Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| |
Collapse
|
9
|
Brandt N, Vierk R, Fester L, Anstötz M, Zhou L, Heilmann LF, Kind S, Steffen P, Rune GM. Sex-specific Difference of Hippocampal Synaptic Plasticity in Response to Sex Neurosteroids. Cereb Cortex 2021; 30:2627-2641. [PMID: 31800024 DOI: 10.1093/cercor/bhz265] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/23/2019] [Accepted: 09/17/2019] [Indexed: 12/14/2022] Open
Abstract
Numerous studies provide increasing evidence, which supports the ideas that every cell in the brain of males may differ from those in females due to differences in sex chromosome complement as well as in response to hormonal effects. In this study, we address the question as to whether actions of neurosteroids, thus steroids, which are synthesized and function within the brain, contribute to sex-specific hippocampal synaptic plasticity. We have previously shown that predominantly in the female hippocampus, does inhibition of the conversion of testosterone to estradiol affect synaptic transmission. In this study, we show that testosterone and its metabolite dihydrotestosterone are essential for hippocampal synaptic transmission specifically in males. This also holds true for the density of mushroom spines and of spine synapses. We obtained similar sex-dependent results using primary hippocampal cultures of male and female animals. Since these cultures originated from perinatal animals, our findings argue for sex-dependent differentiation of hippocampal neurons regarding their responsiveness to sex neurosteroids up to birth, which persist during adulthood. Hence, our in vitro findings may point to a developmental effect either directly induced by sex chromosomes or indirectly by fetal testosterone secretion during the perinatal critical period, when developmental sexual priming takes place.
Collapse
Affiliation(s)
- Nicola Brandt
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ricardo Vierk
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lars Fester
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Max Anstötz
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lepu Zhou
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lukas F Heilmann
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Simon Kind
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Paul Steffen
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
10
|
Cao T, Tang M, Jiang P, Zhang B, Wu X, Chen Q, Zeng C, Li N, Zhang S, Cai H. A Potential Mechanism Underlying the Therapeutic Effects of Progesterone and Allopregnanolone on Ketamine-Induced Cognitive Deficits. Front Pharmacol 2021; 12:612083. [PMID: 33767621 PMCID: PMC7985688 DOI: 10.3389/fphar.2021.612083] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/29/2021] [Indexed: 11/13/2022] Open
Abstract
Ketamine exposure can model cognitive deficits associated with schizophrenia. Progesterone (PROG) and its active metabolite allopregnanolone (ALLO) have neuroprotective effects and the pathway involving progesterone receptor membrane component 1 (PGRMC1), epidermal growth factor receptor (EGFR), glucagon-like peptide-1 receptor (GLP-1R), phosphatidylinositol 3 kinase (PI3K), and protein kinase B (Akt) appears to play a key role in their neuroprotection. The present study aimed to investigate the effects of PROG (8,16 mg kg−1) and ALLO (8,16 mg kg−1) on the reversal of cognitive deficits induced by ketamine (30 mg kg−1) via the PGRMC1 pathway in rat brains, including hippocampus and prefrontal cortex (PFC). Cognitive performance was evaluated by Morris water maze (MWM) test. Western blot and real-time quantitative polymerase chain reaction were utilized to assess the expression changes of protein and mRNA. Additionally, concentrations of PROG and ALLO in plasma, hippocampus and PFC were measured by a liquid chromatography-tandem mass spectrometry method. We demonstrated that PROG or ALLO could reverse the impaired spatial learning and memory abilities induced by ketamine, accompanied with the upregulation of PGRMC1/EGFR/GLP-1R/PI3K/Akt pathway. Additionally, the coadministration of AG205 abolished their neuroprotective effects and induced cognitive deficits similar with ketamine. More importantly, PROG concentrations were markedly elevated in PROG-treated groups in hippocampus, PFC and plasma, so as for ALLO concentrations in ALLO-treated groups. Interestingly, ALLO (16 mg kg−1) significantly increased the levels of PROG. These findings suggest that PROG can exert its neuroprotective effects via activating the PGRMC1/EGFR/GLP-1R/PI3K/Akt pathway in the brain, whereas ALLO also restores cognitive deficits partially via increasing the level of PROG in the brain to activate the PGRMC1 pathway.
Collapse
Affiliation(s)
- Ting Cao
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - MiMi Tang
- Department of Pharmacy, Xiangya Hospital of Central South University, Changsha, China.,Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Pei Jiang
- Institute of Clinical Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - BiKui Zhang
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - XiangXin Wu
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - Qian Chen
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - CuiRong Zeng
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - NaNa Li
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - ShuangYang Zhang
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| | - HuaLin Cai
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
11
|
Maitland NJ. Resistance to Antiandrogens in Prostate Cancer: Is It Inevitable, Intrinsic or Induced? Cancers (Basel) 2021; 13:327. [PMID: 33477370 PMCID: PMC7829888 DOI: 10.3390/cancers13020327] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Increasingly sophisticated therapies for chemical castration dominate first-line treatments for locally advanced prostate cancer. However, androgen deprivation therapy (ADT) offers little prospect of a cure, as resistant tumors emerge rather rapidly, normally within 30 months. Cells have multiple mechanisms of resistance to even the most sophisticated drug regimes, and both tumor cell heterogeneity in prostate cancer and the multiple salvage pathways result in castration-resistant disease related genetically to the original hormone-naive cancer. The timing and mechanisms of cell death after ADT for prostate cancer are not well understood, and off-target effects after long-term ADT due to functional extra-prostatic expression of the androgen receptor protein are now increasingly being recorded. Our knowledge of how these widely used treatments fail at a biological level in patients is deficient. In this review, I will discuss whether there are pre-existing drug-resistant cells in a tumor mass, or whether resistance is induced/selected by the ADT. Equally, what is the cell of origin of this resistance, and does it differ from the treatment-naïve tumor cells by differentiation or dedifferentiation? Conflicting evidence also emerges from studies in the range of biological systems and species employed to answer this key question. It is only by improving our understanding of this aspect of treatment and not simply devising another new means of androgen inhibition that we can improve patient outcomes.
Collapse
Affiliation(s)
- Norman J Maitland
- Department of Biology, University of York, Heslington, York YO10 5DD, UK
| |
Collapse
|
12
|
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: 19] [Impact Index Per Article: 3.8] [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
|
13
|
Dieni CV, Contemori S, Biscarini A, Panichi R. De Novo Synthesized Estradiol: A Role in Modulating the Cerebellar Function. Int J Mol Sci 2020; 21:ijms21093316. [PMID: 32392845 PMCID: PMC7247543 DOI: 10.3390/ijms21093316] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/26/2020] [Accepted: 05/05/2020] [Indexed: 12/29/2022] Open
Abstract
The estrogen estradiol is a potent neuroactive steroid that may regulate brain structure and function. Although the effects of estradiol have been historically associated with gonadal secretion, the discovery that this steroid may be synthesized within the brain has expanded this traditional concept. Indeed, it is accepted that de novo synthesized estradiol in the nervous system (nE2) may modulate several aspects of neuronal physiology, including synaptic transmission and plasticity, thereby influencing a variety of behaviors. These modulations may be on a time scale of minutes via non-classical and often membrane-initiated mechanisms or hours and days by classical actions on gene transcription. Besides the high level, recent investigations in the cerebellum indicate that even a low aromatase expression can be related to the fast nE2 effect on brain functioning. These pieces of evidence point to the importance of an on-demand and localized nE2 synthesis to rapidly contribute to regulating the synaptic transmission. This review is geared at exploring a new scenario for the impact of estradiol on brain processes as it emerges from the nE2 action on cerebellar neurotransmission and cerebellum-dependent learning.
Collapse
Affiliation(s)
- Cristina V. Dieni
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Correspondence: (C.V.D.); (R.P.); Tel.: +1-(205)-996-8660 (C.V.D.); +39-075-5858205 (R.P.)
| | - Samuele Contemori
- Centre for Sensorimotor Performance, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Australia;
| | - Andrea Biscarini
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, 06129 Perugia, Italy;
| | - Roberto Panichi
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, 06129 Perugia, Italy;
- Correspondence: (C.V.D.); (R.P.); Tel.: +1-(205)-996-8660 (C.V.D.); +39-075-5858205 (R.P.)
| |
Collapse
|
14
|
Kohtz AS, Frye CA. Learning and the Lifespan: What's Sex Got to Do With It? Front Neurosci 2020; 14:216. [PMID: 32265631 PMCID: PMC7099170 DOI: 10.3389/fnins.2020.00216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 02/27/2020] [Indexed: 11/23/2022] Open
Abstract
Engagement in sexual behavior can impact neurosteroidogenesis, in particular production of the prohormone testosterone (T) and likely its subsequent metabolism to 5α-androstane-3α-17β-Diol (3α-Diol) or aromatization to estradiol (E2). Androgens and their metabolites vary across the lifespan and impact many behaviors, including cognition, anxiety, and sexual behavior. Thus, we hypothesized that mating may alter cognitive performance via androstane neurosteroids in an age- and experience-dependent manner. We first investigated if exposure to mating during memory consolidation could enhance performance in the novel object recognition task (NOR). Male rats were trained in NOR and then immediately exposed to mating-relevant or control stimuli. Following a 4 h inter-trial interval (ITI), male rats were tested for object memory. Male rats that were exposed to a receptive female during the ITI had better performance in NOR. We then investigated if these effects were due to novelty associated with mating. Male rats were exposed to mating-relevant stimuli and identified as sexually responsive (SR) or sexually non-responsive (SNR) based on a median split of engagement in mating with the stimulus female. We found that a brief history (10 min session daily for five consecutive days) of sexual history substantially influenced performance in the NOR task, such that SR males had better performance in the NOR task, but only when presented with the opportunity to mate during the ITI. As T levels substantially decrease with age in male rodents, we investigated whether the effects of long-term sexual experience (10 months) influenced neurosteroids and NOR performance in mid-aged (12 months old) males. Mid-aged SR males maintain neural T; however, they have decreased neural E2 and decreased cognitive performance at 12 months compared to mid-aged SNR rats. In sexually experienced rats, those with better cognitive performance had greater levels of T metabolites (e.g., 3α-Diol in mated SR males, E2 in mid-aged SNR rats). While naïve males that were mated during the ITI had better cognitive performance, T metabolites were decreased compared to controls. These findings suggest that T metabolites, but not the prohormone, may influence learning dependent on sexual proclivity, experience, and proximate opportunity to mate.
Collapse
Affiliation(s)
- Amy Stave Kohtz
- Department of Psychology, University at Albany – State University of New York (SUNY), Albany, NY, United States
| | - Cheryl A. Frye
- Department of Psychology, University at Albany – State University of New York (SUNY), Albany, NY, United States
- Department of Biological Sciences, University at Albany – State University of New York (SUNY), Albany, NY, United States
- Center for Neuroscience Research, University at Albany – State University of New York (SUNY), Albany, NY, United States
- Center for Life Sciences Research, University at Albany – State University of New York (SUNY), Albany, NY, United States
| |
Collapse
|
15
|
Giatti S, Diviccaro S, Serafini MM, Caruso D, Garcia-Segura LM, Viviani B, Melcangi RC. Sex differences in steroid levels and steroidogenesis in the nervous system: Physiopathological role. Front Neuroendocrinol 2020; 56:100804. [PMID: 31689419 DOI: 10.1016/j.yfrne.2019.100804] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The nervous system, in addition to be a target for steroid hormones, is the source of a variety of neuroactive steroids, which are synthesized and metabolized by neurons and glial cells. Recent evidence indicates that the expression of neurosteroidogenic proteins and enzymes and the levels of neuroactive steroids are different in the nervous system of males and females. We here summarized the state of the art of neuroactive steroids, particularly taking in consideration sex differences occurring in the synthesis and levels of these molecules. In addition, we discuss the consequences of sex differences in neurosteroidogenesis for the function of the nervous system under healthy and pathological conditions and the implications of neuroactive steroids and neurosteroidogenesis for the development of sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Melania Maria Serafini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Barbara Viviani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
16
|
Finney CA, Shvetcov A, Westbrook RF, Jones NM, Morris MJ. The role of hippocampal estradiol in synaptic plasticity and memory: A systematic review. Front Neuroendocrinol 2020; 56:100818. [PMID: 31843506 DOI: 10.1016/j.yfrne.2019.100818] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/29/2019] [Accepted: 12/11/2019] [Indexed: 12/31/2022]
Abstract
The consolidation of long-term memory is influenced by various neuromodulators. One of these is estradiol, a steroid hormone that is synthesized both in peripheral endocrine tissue and in the brain, including the hippocampus. Here, we examine the evidence regarding the role of estradiol in the hippocampus, specifically, in memory formation and its effects on the molecular mechanisms underlying synaptic plasticity. We conclude that estradiol improves memory consolidation and, thereby, long-term memory. Previous studies have shown that it does this in three, interconnected ways: (1) via functional changes in excitatory activity, (2) signaling changes in calcium dynamics, protein phosphorylation and protein expression, and (3) structural changes to synaptic morphology. Through a functional network analysis of proteins affected by estradiol, we identify potential protein-protein interactions that further support a role for estradiol in modulating synaptic plasticity as well as highlight signaling pathways that may be involved in these changes within the hippocampus.
Collapse
Affiliation(s)
- C A Finney
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - A Shvetcov
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - R F Westbrook
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - N M Jones
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - M J Morris
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
17
|
Çalışkan G, Raza SA, Demiray YE, Kul E, Sandhu KV, Stork O. Depletion of dietary phytoestrogens reduces hippocampal plasticity and contextual fear memory stability in adult male mouse. Nutr Neurosci 2019; 24:951-962. [PMID: 31814540 DOI: 10.1080/1028415x.2019.1698826] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Phytoestrogens are non-steroidal estrogen analogues and are found primarily in soy products. They have received increasing attention as dietary supplements for estrogen deficiency and as modulators of endogenous estrogen functions, including cognition and emotion. In addition to modifying the levels of circulating sex hormones, phytoestrogens also exert direct effects on estrogen and androgen receptors in the brain and thus effectively modulate the neural circuit functions.Objective: The aim of this study was to investigate the long-term effects of low phytoestrogen intake (∼6 weeks) on the hippocampal plasticity and hippocampus-dependent memory formation in the adult C57BL/6 male mice.Methods and Results: In comparison to mice on a diet with normal phytoestrogen content, mice on low phytoestrogen diet showed a significant reduction in the phosphorylation of NR2B subunit, a molecular correlate of plasticity in the Schaffer collateral-CA1 synapse. We observed a profound decrease in long-term potentiation (LTP) in the ventral hippocampus, whereas no effect on plasticity was evident in its dorsal portion. Furthermore, we demonstrated that acute perfusion of slices with an estrogen analogue equol, an isoflovane metabolized from daidzein produced by the bacterial flora in the gut, was able to rescue the observed LTP deficit. Examining potential behavioral correlates of the plasticity attenuation, we found that mice on phytoestrogen-free diet display decreased contextual fear memory at remote but not at recent time points after training.Conclusions: Our data suggests that nutritional phytoestrogens have profound effects on the plasticity in the ventral hippocampus and ventral hippocampus-dependent memory.
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Syed Ahsan Raza
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Yunus E Demiray
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Emre Kul
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Kiran V Sandhu
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Oliver Stork
- Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| |
Collapse
|
18
|
Tozzi A, Durante V, Manca P, Di Mauro M, Blasi J, Grassi S, Calabresi P, Kawato S, Pettorossi VE. Bidirectional Synaptic Plasticity Is Driven by Sex Neurosteroids Targeting Estrogen and Androgen Receptors in Hippocampal CA1 Pyramidal Neurons. Front Cell Neurosci 2019; 13:534. [PMID: 31866827 PMCID: PMC6904272 DOI: 10.3389/fncel.2019.00534] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/18/2019] [Indexed: 11/23/2022] Open
Abstract
Neuroactive estrogenic and androgenic steroids influence synaptic transmission, finely modulating synaptic plasticity in several brain regions including the hippocampus. While estrogens facilitate long-term potentiation (LTP), androgens are involved in the induction of long-term depression (LTD) and depotentiation (DP) of synaptic transmission. To examine sex neurosteroid-dependent LTP and LTD in single cells, patch-clamp recordings from hippocampal CA1 pyramidal neurons of male rats and selective antagonists for estrogen receptors (ERs) and androgen (AR) receptors were used. LTP induced by high-frequency stimulation (HFS) depended on activation of ERs since it was prevented by the ER antagonist ICI 182,780 in most of the neurons. Application of the selective antagonists for ERα (MPP) or ERβ (PHTPP) caused a reduction of the LTP amplitude, while these antagonists in combination, prevented LTP completely. LTP was never affected by blocking AR with the specific antagonist flutamide. Conversely, LTD and DP, elicited by low-frequency stimulation (LFS), were impeded by flutamide, but not by ICI 182,780, in most neurons. In few cells, LTD was even reverted to LTP by flutamide. Moreover, the combined application of both ER and AR antagonists completely prevented both LTP and LTD/DP in the same neuron. The current study demonstrates that the activation of ERs is necessary for inducing LTP in hippocampal pyramidal neurons, whereas the activation of ARs is required for LTD and DP. Moreover, both estrogen- and androgen-dependent LTP and LTD can be expressed in the same pyramidal neurons, suggesting that the activation of sex neurosteroids signaling pathways is responsible for bidirectional synaptic plasticity.
Collapse
Affiliation(s)
- Alessandro Tozzi
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, Perugia, Italy
| | - Valentina Durante
- Department of Medicine, Section of Neurological Clinic, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Paolo Manca
- Department de Patologia i Terapèutica Experimental, Facultat de Medicina, Campus de Bellvitge, Universitat de Barcelona, Barcelona, Spain
| | - Michela Di Mauro
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, Perugia, Italy
| | - Juan Blasi
- Department de Patologia i Terapèutica Experimental, Facultat de Medicina, Campus de Bellvitge, Universitat de Barcelona, Barcelona, Spain
| | - Silvarosa Grassi
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, Perugia, Italy
| | - Paolo Calabresi
- Department of Medicine, Section of Neurological Clinic, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Suguru Kawato
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan.,Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Vito Enrico Pettorossi
- Department of Experimental Medicine, Section of Physiology and Biochemistry, University of Perugia, Perugia, Italy
| |
Collapse
|
19
|
Neuron-Derived Estrogen Regulates Synaptic Plasticity and Memory. J Neurosci 2019; 39:2792-2809. [PMID: 30728170 PMCID: PMC6462452 DOI: 10.1523/jneurosci.1970-18.2019] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 12/28/2018] [Accepted: 01/18/2019] [Indexed: 01/27/2023] Open
Abstract
17β-estradiol (E2) is produced from androgens via the action of the enzyme aromatase. E2 is known to be made in neurons in the brain, but its precise functions in the brain are unclear. Here, we used a forebrain-neuron-specific aromatase knock-out (FBN-ARO-KO) mouse model to deplete neuron-derived E2 in the forebrain of mice and thereby elucidate its functions. FBN-ARO-KO mice showed a 70–80% decrease in aromatase and forebrain E2 levels compared with FLOX controls. Male and female FBN-ARO-KO mice exhibited significant deficits in forebrain spine and synaptic density, as well as hippocampal-dependent spatial reference memory, recognition memory, and contextual fear memory, but had normal locomotor function and anxiety levels. Reinstating forebrain E2 levels via exogenous in vivo E2 administration was able to rescue both the molecular and behavioral defects in FBN-ARO-KO mice. Furthermore, in vitro studies using FBN-ARO-KO hippocampal slices revealed that, whereas induction of long-term potentiation (LTP) was normal, the amplitude was significantly decreased. Intriguingly, the LTP defect could be fully rescued by acute E2 treatment in vitro. Mechanistic studies revealed that FBN-ARO-KO mice had compromised rapid kinase (AKT, ERK) and CREB-BDNF signaling in the hippocampus and cerebral cortex. In addition, acute E2 rescue of LTP in hippocampal FBN-ARO-KO slices could be blocked by administration of a MEK/ERK inhibitor, further suggesting a key role for rapid ERK signaling in neuronal E2 effects. In conclusion, the findings provide evidence of a critical role for neuron-derived E2 in regulating synaptic plasticity and cognitive function in the male and female brain. SIGNIFICANCE STATEMENT The steroid hormone 17β-estradiol (E2) is well known to be produced in the ovaries in females. Intriguingly, forebrain neurons also express aromatase, the E2 biosynthetic enzyme, but the precise functions of neuron-derived E2 is unclear. Using a novel forebrain-neuron-specific aromatase knock-out mouse model to deplete neuron-derived E2, the current study provides direct genetic evidence of a critical role for neuron-derived E2 in the regulation of rapid AKT-ERK and CREB-BDNF signaling in the mouse forebrain and demonstrates that neuron-derived E2 is essential for normal expression of LTP, synaptic plasticity, and cognitive function in both the male and female brain. These findings suggest that neuron-derived E2 functions as a novel neuromodulator in the forebrain to control synaptic plasticity and cognitive function.
Collapse
|
20
|
Hajali V, Andersen ML, Negah SS, Sheibani V. Sex differences in sleep and sleep loss-induced cognitive deficits: The influence of gonadal hormones. Horm Behav 2019; 108:50-61. [PMID: 30597139 DOI: 10.1016/j.yhbeh.2018.12.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 12/23/2018] [Accepted: 12/25/2018] [Indexed: 12/30/2022]
Abstract
Males and females can respond differentially to the same environmental stimuli and experimental conditions. Chronic sleep loss is a frequent and growing problem in many modern societies and has a broad variety of negative outcomes for health and well-being. While much has been done to explore the deleterious effects of sleep deprivation (SD) on cognition in both human and animal studies over the last few decades, very little attention has been paid to the part played by sex differences and gonadal steroids in respect of changes in cognitive functions caused by sleep loss. The effects of gonadal hormones on sleep regulation and cognitive performances are well established. Reduced gonadal function in menopausal women and elderly men is associated with sleep disturbances and cognitive decline as well as dementia, which suggests that sex steroids play a key role in modulating these conditions. Finding out whether there are sex differences in respect of the effect of insufficient sleep on cognition, and how neuroendocrine mediators influence cognitive impairment induced by SD could provide valuable insights into the best therapies for each sex. In this review, we aim to highlight the involvement of sex differences and gonadal hormone status on the severity of cognitive deficits induced by sleep deficiency in both human and animal studies.
Collapse
Affiliation(s)
- Vahid Hajali
- Department of Neuroscience, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Monica L Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo - UNIFESP, Brazil
| | - Sajad Sahab Negah
- Department of Neuroscience, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
21
|
Neural-derived estradiol regulates brain plasticity. J Chem Neuroanat 2018; 89:53-59. [DOI: 10.1016/j.jchemneu.2017.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/16/2017] [Accepted: 04/12/2017] [Indexed: 01/12/2023]
|
22
|
Yang Y, Fang Z, Dai Y, Wang Y, Liang Y, Zhong X, Wang Q, Hu Y, Zhang Z, Wu D, Xu X. Bisphenol-A antagonizes the rapidly modulating effect of DHT on spinogenesis and long-term potentiation of hippocampal neurons. CHEMOSPHERE 2018; 195:567-575. [PMID: 29278848 DOI: 10.1016/j.chemosphere.2017.12.086] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 06/07/2023]
Abstract
Bisphenol A (BPA), a common environmental endocrine disruptor, modulates estrogenic, antiestrogenic, and antiandrogenic effects throughout the lifespan. Recent studies found more obvious adverse effect of BPA on some neurobehavior in males than that in females. In this study, BPA at 10-100 nM rapidly increased the densities of the dendrite spine and synapse in cultured hippocampal neurons of rats in vitro within 1 h. Co-treatment of BPA (100 nM) with dihydrotestosterone (DHT, 10 nM) or with 17β-E2 (10 nM) completely eliminated the promotion of DHT or 17β-E2 in the densities of the dendritic spine and synapse. Pretreatment of estrogen receptors (ERs) antagonist ICI182,780 but not of androgen receptors (ARs) antagonist flutamide (Flu) for 30min completely blocked BPA-enhanced densities of the dendritic spine and synapse. Pretreatment of flutamide for 30min before BPA and DHT completely rescued BPA-enhanced densities of the dendritic spine and synapse. Furthermore, pretreatment of ERK1/2 inhibitor U0126 or p38 inhibitor SB203580 entirely eliminated BPA-induced increases in the densities of the dendritic spine and synapse. Meanwhile, BPA (100 nM) enhanced long-term potentiation (LTP) induction of dentate gyrus in hippocampal slices of younger male rats, which was not blocked by co-incubation of flutamide but was inhibited by pretreatment of an P38 inhibitor SB203580. Co-application of BPA with DHT inhibited DHT-suppressed LTP. These results are the first demonstrating the antagonism of BPA to the rapid modification of DHT in synaptic plasticity. However, BPA alone rapidly promotes spinogenesis and synaptic activity through ER instead of AR, and both ERKs and p38 signaling pathways are involved in these processes.
Collapse
Affiliation(s)
- Yang Yang
- Chemistry and Life Sciences College, Key Laboratory of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, PR China
| | - Zhaoqing Fang
- Chemistry and Life Sciences College, Key Laboratory of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, PR China
| | - Yuhua Dai
- Chemistry and Life Sciences College, Key Laboratory of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, PR China
| | - Yu Wang
- Chemistry and Life Sciences College, Key Laboratory of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, PR China
| | - Yufeng Liang
- Chemistry and Life Sciences College, Key Laboratory of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, PR China
| | - Xiaoyu Zhong
- Chemistry and Life Sciences College, Key Laboratory of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, PR China
| | - Qinwen Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, PR China
| | - Yizhong Hu
- Chemistry and Life Sciences College, Key Laboratory of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, PR China
| | - Zigui Zhang
- Zhejiang Provincial Key Laboratory of Ecology, Xingzhi College, Zhejiang Normal University, PR China
| | - Donghong Wu
- Chemistry and Life Sciences College, Key Laboratory of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, PR China
| | - Xiaohong Xu
- Chemistry and Life Sciences College, Key Laboratory of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, PR China.
| |
Collapse
|
23
|
Murakami G, Hojo Y, Kato A, Komatsuzaki Y, Horie S, Soma M, Kim J, Kawato S. Rapid nongenomic modulation by neurosteroids of dendritic spines in the hippocampus: Androgen, oestrogen and corticosteroid. J Neuroendocrinol 2018; 30. [PMID: 29194818 DOI: 10.1111/jne.12561] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/22/2022]
Abstract
Memories are stored in synapses that consist of axon terminals and dendritic spines. Dendritic spines are postsynaptic structures of synapses and are essential for synaptic plasticity and cognition. Therefore, extensive investigations concerning the functions and structures of spines have been performed. Sex steroids and stress steroids have been shown to modulate hippocampal synapses. Although the rapid modulatory action of sex steroids on synapses has been studied in hippocampal neurones over several decades, the essential molecular mechanisms have not been fully understood. Here, a description of kinase-dependent signalling mechanisms is provided that can explain the rapid nongenomic modulation of dendritic spinogenesis in rat and mouse hippocampal slices by the application of sex steroids, including dihydrotestosterone, testosterone, oestradiol and progesterone. We also indicate the role of synaptic (classic) sex steroid receptors that trigger these rapid synaptic modulations. Moreover, we describe rapid nongenomic spine modulation by applying corticosterone, which is an acute stress model of the hippocampus. The explanations for the results obtained are mainly based on the optical imaging of dendritic spines. Comparisons are also performed with results obtained from other types of imaging, including electron microscopic imaging. Relationships between spine modulation and modulation of cognition are discussed. We recognise that most of rapid effects of exogenously applied oestrogen and androgen were observed in steroid-depleted conditions, including acute slices of the hippocampus, castrated male animals and ovariectomised female animals. Therefore, the previously observed effects can be considered as a type of recovery event, which may be essentially similar to hormone replacement therapy under hormone-decreased conditions. On the other hand, in gonadally intact young animals with high levels of endogenous sex hormones, further supplementation of sex hormones might not be effective, whereas the infusion of blockers for steroid receptors or kinases may be effective, with respect to suppressing sex hormone functions, thus providing useful information regarding molecular mechanisms.
Collapse
Affiliation(s)
- G Murakami
- Department of Liberal Arts, Faculty of Medicine, Saitama Medical University, Iruma, Saitama, Japan
| | - Y Hojo
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Iruma, Saitama, Japan
| | - A Kato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Meguro, Tokyo, Japan
| | - Y Komatsuzaki
- Department of Physics, College of Science and Technology, Nihon University, Chiyoda, Tokyo, Japan
| | - S Horie
- Department of Urology, Graduate School of Medicine, Juntendo University, Hongo, Tokyo, Japan
| | - M Soma
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Itabashi, Tokyo, Japan
| | - J Kim
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Itabashi, Tokyo, Japan
| | - S Kawato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Meguro, Tokyo, Japan
- Department of Urology, Graduate School of Medicine, Juntendo University, Hongo, Tokyo, Japan
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Itabashi, Tokyo, Japan
| |
Collapse
|
24
|
Hojo Y, Kawato S. Neurosteroids in Adult Hippocampus of Male and Female Rodents: Biosynthesis and Actions of Sex Steroids. Front Endocrinol (Lausanne) 2018; 9:183. [PMID: 29740398 PMCID: PMC5925962 DOI: 10.3389/fendo.2018.00183] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
The brain is not only the target of steroid hormones but also is able to locally synthesize steroids de novo. Evidence of the local production of steroids in the brain has been accumulating in various vertebrates, including teleost fish, amphibia, birds, rodents, non-human primates, and humans. In this review, we mainly focus on the local production of sex steroids in the hippocampal neurons of adult rodents (rats and mice), a center for learning and memory. From the data of the hippocampus of adult male rats, hippocampal principal neurons [pyramidal cells in CA1-CA3 and granule cells in dentate gyrus (DG)] have a complete system for biosynthesis of sex steroids. Liquid chromatography with tandem-mass-spectrometry (LC-MS/MS) enabled us to accurately determine the levels of hippocampal sex steroids including 17β-estradiol (17β-E2), testosterone (T), and dihydrotestosterone (DHT), which are much higher than those in blood. Next, we review the steroid synthesis in the hippocampus of female rats, since previous knowledge had been biased toward the data from males. Recently, we clarified that the levels of hippocampal steroids fluctuate in adult female rats across the estrous cycle. Accurate determination of hippocampal steroids at each stage of the estrous cycle is of importance for providing the account for the fluctuation of female hippocampal functions, including spine density, long-term potentiation (LTP) and long-term depression (LTD), and learning and memory. These functional fluctuations in female had been attributed to the level of circulation-derived steroids. LC-MS/MS analysis revealed that the dendritic spine density in CA1 of adult female hippocampus correlates with the levels of hippocampal progesterone and 17β-E2. Finally, we introduce the direct evidence of the role of hippocampus-synthesized steroids in hippocampal function including neurogenesis, LTP, and memory consolidation. Mild exercise (2 week of treadmill running) elevated synthesis of DHT in the hippocampus, but not in the testis, of male rats, resulting in enhancement of neurogenesis in DG. Concerning synaptic plasticity, hippocampus-synthesized E2 is required for LTP induction, whereas hippocampus-synthesized DHT is required for LTD induction. Furthermore, hippocampus-synthesized E2 is involved in memory consolidation tested by object recognition and object placement tasks, both of which are hippocampus-dependent.
Collapse
Affiliation(s)
- Yasushi Hojo
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
- *Correspondence: Yasushi Hojo,
| | - Suguru Kawato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Cognitive Neuroscience, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| |
Collapse
|
25
|
Pinar C, Fontaine CJ, Triviño-Paredes J, Lottenberg CP, Gil-Mohapel J, Christie BR. Revisiting the flip side: Long-term depression of synaptic efficacy in the hippocampus. Neurosci Biobehav Rev 2017. [PMID: 28624435 DOI: 10.1016/j.neubiorev.2017.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Synaptic plasticity is widely regarded as a putative biological substrate for learning and memory processes. While both decreases and increases in synaptic strength are seen as playing a role in learning and memory, long-term depression (LTD) of synaptic efficacy has received far less attention than its counterpart long-term potentiation (LTP). Never-the-less, LTD at synapses can play an important role in increasing computational flexibility in neural networks. In addition, like learning and memory processes, the magnitude of LTD can be modulated by factors that include stress and sex hormones, neurotrophic support, learning environments, and age. Examining how these factors modulate hippocampal LTD can provide the means to better elucidate the molecular underpinnings of learning and memory processes. This is in turn will enhance our appreciation of how both increases and decreases in synaptic plasticity can play a role in different neurodevelopmental and neurodegenerative conditions.
Collapse
Affiliation(s)
- Cristina Pinar
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Christine J Fontaine
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Juan Triviño-Paredes
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Carina P Lottenberg
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada; Faculty of Medical Sciences of Santa Casa de São Paulo, Sao Paulo, SP, Brazil
| | - Joana Gil-Mohapel
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Brian R Christie
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada.
| |
Collapse
|
26
|
Di Mauro M, Tozzi A, Calabresi P, Pettorossi VE, Grassi S. Different synaptic stimulation patterns influence the local androgenic and estrogenic neurosteroid availability triggering hippocampal synaptic plasticity in the male rat. Eur J Neurosci 2017; 45:499-509. [DOI: 10.1111/ejn.13455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/21/2016] [Accepted: 10/27/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Michela Di Mauro
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
| | - Alessandro Tozzi
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
- Fondazione Santa Lucia – I.R.C.C.S. Rome Italy
| | - Paolo Calabresi
- Fondazione Santa Lucia – I.R.C.C.S. Rome Italy
- Dipartimento di Medicina Clinica Neurologica Università di Perugia Perugia Italy
| | - Vito Enrico Pettorossi
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
| | - Silvarosa Grassi
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
| |
Collapse
|
27
|
Sex-Dependent Regulation of Aromatase-Mediated Synaptic Plasticity in the Basolateral Amygdala. J Neurosci 2016; 37:1532-1545. [PMID: 28028198 DOI: 10.1523/jneurosci.1532-16.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 11/11/2016] [Accepted: 12/11/2016] [Indexed: 02/01/2023] Open
Abstract
The basolateral amygdala (BLA) integrates sensory input from cortical and subcortical regions, a function that requires marked synaptic plasticity. Here we provide evidence that cytochrome P450 aromatase (AROM), the enzyme converting testosterone to 17β-estradiol (E2), contributes to the regulation of this plasticity in a sex-specific manner. We show that AROM is expressed in the BLA, particularly in the basolateral nucleus (BL), in male and female rodents. Systemic administration of the AROM inhibitor letrozole reduced spine synapse density in the BL of adult female mice but not in the BL of male mice. Similarly, in organotypic corticoamygdalar slice cultures from immature rats, treatment with letrozole significantly reduced spine synapses in the BL only in cultures derived from females. In addition, letrozole sex-specifically altered synaptic properties in the BL: in acute slices from juvenile (prepubertal) female rats, wash-in of letrozole virtually abolished long-term potentiation (LTP), whereas it did not prevent the generation of LTP in the slices from males. Together, these data indicate that neuron-derived E2 modulates synaptic plasticity in rodent BLA sex-dependently. As protein expression levels of AROM, estrogen and androgen receptors did not differ between males and females and were not sex-specifically altered by letrozole, the findings suggest sex-specific mechanisms of E2 signaling.SIGNIFICANCE STATEMENT The basolateral amygdala (BLA) is a key structure of the fear circuit. This research reveals a sexually dimorphic regulation of synaptic plasticity in the BLA involving neuronal aromatase, which produces the neurosteroid 17β-estradiol (E2). As male and female neurons in rodent BLA responded differently to aromatase inhibition both in vivo and in vitro, our findings suggest that E2 signaling in BLA neurons is regulated sex-dependently, presumably via mechanisms that have been established during sexual determination. These findings could be relevant for the understanding of sex differences in mood disorders and of the side effects of cytochrome P450 aromatase inhibitors, which are frequently used for breast cancer therapy.
Collapse
|