1
|
Sun Q, Li G, Zhao F, Dong M, Xie W, Liu Q, Yang W, Cui R. Role of estrogen in treatment of female depression. Aging (Albany NY) 2024; 16:3021-3042. [PMID: 38309292 PMCID: PMC10911346 DOI: 10.18632/aging.205507] [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: 07/18/2023] [Accepted: 11/28/2023] [Indexed: 02/05/2024]
Abstract
Depression is a neurological disorder that profoundly affects human physical and mental health, resulting in various changes in the central nervous system. Despite several prominent hypotheses, such as the monoaminergic theory, hypothalamic-pituitary-adrenal (HPA) axis theory, neuroinflammation, and neuroplasticity, the current understanding of depression's pathogenesis remains incomplete. Importantly, depression is a gender-dimorphic disorder, with women exhibiting higher incidence rates than men. Given estrogen's pivotal role in the menstrual cycle, it is reasonable to postulate that its fluctuating levels could contribute to the pathogenesis of depression. Estrogen acts by binding to a diversity of receptors, which are widely distributed in the central nervous system. An abundance of research has established that estrogen and its receptors play a crucial role in depression, spanning pathogenesis and treatment. In this comprehensive review, we provide an in-depth analysis of the fundamental role of estrogen and its receptors in depression, with a focus on neuroinflammation, neuroendocrinology, and neuroplasticity. Furthermore, we discuss potential mechanisms underlying the therapeutic effects of estrogen in the treatment of depression, which may pave the way for new antidepressant drug development and alternative treatment options.
Collapse
Affiliation(s)
- Qihan Sun
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Guangquan Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Mengmeng Dong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Wei Xie
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Qianqian Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Wei Yang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
2
|
Sheppard PAS, Chandramohan D, Lumsden A, Vellone D, Denley MCS, Srivastava DP, Choleris E. Social memory in female mice is rapidly modulated by 17β-estradiol through ERK and Akt modulation of synapse formation. Proc Natl Acad Sci U S A 2023; 120:e2300191120. [PMID: 37490537 PMCID: PMC10400940 DOI: 10.1073/pnas.2300191120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/25/2023] [Indexed: 07/27/2023] Open
Abstract
Social memory is essential to the functioning of a social animal within a group. Estrogens can affect social memory too quickly for classical genomic mechanisms. Previously, 17β-estradiol (E2) rapidly facilitated short-term social memory and increased nascent synapse formation, these synapses being potentiated following neuronal activity. However, what mechanisms underlie and coordinate the rapid facilitation of social memory and synaptogenesis are unclear. Here, the necessity of extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K) signaling for rapid facilitation of short-term social memory and synaptogenesis was tested. Mice performed a short-term social memory task or were used as task-naïve controls. ERK and PI3K pathway inhibitors were infused intradorsal hippocampally 5 min before E2 infusion. Forty minutes following intrahippocampal E2 or vehicle administration, tissues were collected for quantification of glutamatergic synapse number in the CA1. Dorsal hippocampal E2 rapid facilitation of short-term social memory depended upon ERK and PI3K pathways. E2 increased glutamatergic synapse number (bassoon puncta positive for GluA1) in task-performing mice but decreased synapse number in task-naïve mice. Critically, ERK signaling was required for synapse formation/elimination in task-performing and task-naïve mice, whereas PI3K inhibition blocked synapse formation only in task-performing mice. While ERK and PI3K are both required for E2 facilitation of short-term social memory and synapse formation, only ERK is required for synapse elimination. This demonstrates previously unknown, bidirectional, rapid actions of E2 on brain and behavior and underscores the importance of estrogen signaling in the brain to social behavior.
Collapse
Affiliation(s)
- Paul A. S. Sheppard
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ONN1G 2W1, Canada
| | - Deepthi Chandramohan
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, LondonWC2R 2LS, United Kingdom
- Medical Research Council Centre for Neurodevelopmental Disorders, King’s College London, LondonWC2R 2LS, United Kingdom
| | - Alanna Lumsden
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ONN1G 2W1, Canada
| | - Daniella Vellone
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ONN1G 2W1, Canada
| | - Matthew C. S. Denley
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, LondonWC2R 2LS, United Kingdom
- Medical Research Council Centre for Neurodevelopmental Disorders, King’s College London, LondonWC2R 2LS, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, LondonWC2R 2LS, United Kingdom
- Medical Research Council Centre for Neurodevelopmental Disorders, King’s College London, LondonWC2R 2LS, United Kingdom
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ONN1G 2W1, Canada
| |
Collapse
|
3
|
Anti-Inflammatory Actions of G-Protein-Coupled Estrogen Receptor 1 (GPER) and Brain-Derived Estrogen Following Cerebral Ischemia in Ovariectomized Rats. BIOLOGY 2023; 12:biology12010099. [PMID: 36671793 PMCID: PMC9855882 DOI: 10.3390/biology12010099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Global cerebral ischemia can elicit rapid innate neuroprotective mechanisms that protect against delayed neuronal death. Brain-derived 17β-estradiol (BDE2), an endogenous neuroprotectant, is synthesized from testosterone by the enzyme aromatase (Aro) and is upregulated by brain ischemia and inflammation. Our recent study revealed that G1, a specific G-protein-coupled estrogen receptor 1 (GPER) agonist, exerts anti-inflammatory and anti-apoptotic roles after global cerebral ischemia (GCI). Herein, we aimed to elucidate whether G1 modulates the early inflammatory process and the potential underlying mechanisms in the ovariectomized rat hippocampal CA1 region. G1 was found to markedly reduce pro-inflammatory (iNOS, MHCII, and CD68) and to enhance anti-inflammatory (CD206, Arginase 1, IL1RA, PPARγ, and BDNF) markers after 1 and 3 days of reperfusion after GCI. Intriguingly, the neuroprotection of G1 was blocked by the Aro inhibitor, letrozole. Conversely, the GPER antagonist, G36, inhibited Aro-BDE2 signaling and exacerbated neuronal damage. As a whole, this work demonstrates a novel anti-inflammatory role of GPER, involving a synergistic mediation with BDE2 during the early stage of GCI.
Collapse
|
4
|
Eroğlu İ, Eroğlu BÇ. Potential role of tryptophan catabolism in cancer-related cognitive impairment. Nutrition 2022; 103-104:111765. [PMID: 35908496 DOI: 10.1016/j.nut.2022.111765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/31/2022] [Indexed: 12/24/2022]
Abstract
Oncology may be the most rapidly expanding field in medicine, with several innovative diagnostic and therapeutic procedures appearing daily. Advances in oncology have improved the survival rate for patients with cancer and promoting quality of life is now one of the goals in the care of these patients. Patients face a variety of disease- and treatment-related side effects, including anorexia, nausea, vomiting, recurring infections, and sleep difficulties. Cancer-related cognitive impairment (CRCI) is an overlooked clinical condition found in oncologic practice, particularly in patients with breast cancer. Although several potential mechanisms for CRCI have been hypothesized, to our knowledge, the exact mechanism is still unknown. Alterations in the tryptophan kynurenine pathway have been shown to impair cognitive skills in several mental illnesses. However, its possible function in CRCI has yet to be investigated. The aim of this was to examine the possible interactions between tryptophan catabolism and CRCI.
Collapse
Affiliation(s)
- İmdat Eroğlu
- Hacettepe University Faculty of Medicine, Department of Internal Medicine, Ankara, Turkey.
| | - Burcu Çelik Eroğlu
- Hacettepe University Faculty of Medicine, Department of Internal Medicine, Ankara, Turkey
| |
Collapse
|
5
|
Wen Z, Hammoud MZ, Scott JC, Jimmy J, Brown L, Marin MF, Asnaani A, Gur RC, Foa EB, Milad MR. Impact of exogenous estradiol on task-based and resting-state neural signature during and after fear extinction in healthy women. Neuropsychopharmacology 2021; 46:2278-2287. [PMID: 34493827 PMCID: PMC8581031 DOI: 10.1038/s41386-021-01158-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023]
Abstract
Fluctuations of endogenous estrogen modulates fear extinction, but the influence of exogenous estradiol is less studied. Moreover, little focus has been placed on the impact of estradiol on broad network connectivity beyond the fear extinction circuit. Here, we examined the effect of acute exogenous estradiol administration on fear extinction-induced brain activation, whole-brain functional connectivity (FC) during the fear extinction task and post-extinction resting-state. Ninety healthy women (57 using oral contraceptives [OC], 33 naturally cycling [NC]) were fear conditioned on day 1. They ingested an estradiol or placebo pill prior to extinction learning on day 2 (double-blind design). Extinction memory was assessed on day 3. Task-based functional MRI data were ascertained on days 2 and 3 and resting-state data were collected post-extinction on day 2 and pre-recall on day 3. Estradiol administration significantly modulated the neural signature associated with fear extinction learning and memory, consistent with prior studies. Importantly, estradiol administration induced significant changes in FC within multiple networks, including the default mode and somatomotor networks during extinction learning, post-extinction, and during extinction memory recall. Exploratory analyses revealed that estradiol impacted ventromedial prefrontal cortex (vmPFC) activation and FC differently in the NC and OC women. The data implicate a more diffused and significant effect of acute estradiol administration on multiple networks. Such an effect might be beneficial to modulating attention and conscious processes in addition to engaging neural processes associated with emotional learning and memory consolidation.
Collapse
Affiliation(s)
- Zhenfu Wen
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Mira Z Hammoud
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - J Cobb Scott
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- VISN4 Mental Illness Research, Education, and Clinical Center at the Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - Jagan Jimmy
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Lily Brown
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marie-France Marin
- Departement of Psychology, Université du Québec à Montréal, Montreal, QC, Canada
- Research Center of the Institut universitaire en santé mentale de Montréal, Montreal, QC, Canada
| | - Anu Asnaani
- Department of Psychology, University of Utah, Salt Lake City, UT, USA
| | - Ruben C Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edna B Foa
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohammed R Milad
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
- The Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| |
Collapse
|
6
|
Endocrine Therapy With or Without CDK4/6 Inhibitors in Women With Hormone-receptor Positive Breast Cancer: What do we Know About the Effects on Cognition? Clin Breast Cancer 2021; 22:191-199. [PMID: 34556423 DOI: 10.1016/j.clbc.2021.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/01/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022]
Abstract
Adjuvant endocrine therapy (ET) is the cornerstone of treatment for hormone-receptor positive breast cancer. Recently, ET is increasingly combined with "cyclin-dependent kinases 4 and 6'' (CDK4/6) inhibitors. Given the importance of estrogens in neural processes and the role of cyclin D in hippocampal cell proliferation, it is plausible that these therapies affect cognition, but studies on these potential cognitive effects are sparse. In this review, we summarize existing knowledge on the cognitive effects of ET and CDK4/6 inhibitors in pre-, peri- and postmenopausal patients with breast cancer. We show that several clinical studies support adverse cognitive effects, especially on verbal memory, after ET-induced decrease of estrogen-levels or inactivation of estrogen-receptors. Clinical studies on the cognitive effects of CDK4/6 inhibitors are virtually non-existent and no conclusions can yet be drawn. Longitudinal studies on the cognitive effects of the combined ET-CDK4/6 inhibitors are highly needed to properly inform patients about potential short-term and long-term cognitive side effects. These studies should preferably include cognitive assessments (including a measurement prior to ET), and be designed in such a way that they can account for variables such as type and duration of ET, CDK4/6 inhibition, menopausal status, and other disease- and treatment-related symptoms that can impact cognition, such as fatigue and distress.
Collapse
|
7
|
Barok R, Grittner JML, Dougherty BJ. The long-term impact of ovariectomy on ventilation and expression of phrenic long-term facilitation in female rats. Exp Physiol 2021; 106:2002-2012. [PMID: 34180081 PMCID: PMC8410681 DOI: 10.1113/ep089546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/18/2021] [Indexed: 01/04/2023]
Abstract
NEW FINDINGS What is the central question of this study? Would ovariectomy cause prolonged changes in ventilation and sustained loss of acute, intermittent hypoxia-induced neuroplasticity or would these outcomes be restored with time? What is the main finding and its importance? Our main findings demonstrate that ovariectomy elicits minimal alteration in overall breathing function but impairs acute, intermittent hypoxia-induced plasticity for ≤ 12 weeks. ABSTRACT Sex hormones are necessary to enable respiratory neuroplasticity, including phrenic long-term facilitation (pLTF), a form of respiratory motor plasticity elicited by acute, intermittent hypoxia (AIH). Female rats exhibit a progressive increase in phrenic nerve amplitude after AIH characteristic of pLTF only during pro-oestrus, the stage of the oestrous cycle notable for elevated circulating oestradiol levels. Removal of the ovaries [ovariectomy (OVX)], the primary source of circulating oestradiol, also eliminates AIH-induced pLTF after 1 week. Ovariectomy is used routinely as a model to examine the impact of sex hormones on CNS structure and function, but the long-term impact of OVX is rarely examined. Extra-ovarian sites of oestradiol synthesis, including multiple CNS sites, have been identified and might possess the capacity to restore oestradiol levels, in part, over time, impacting respiratory function and the expression of respiratory neuroplasticity. We examined both ventilation in awake, freely behaving female rats, using barometric plethysmography, and the expression of AIH-induced pLTF in anaesthetized, ventilated female rats 2 and 12 weeks after OVX and compared them with age-matched ovarian-intact female rats. Our findings indicate that chronic OVX had little impact on baseline breathing or in the response to respiratory challenge (10% O2 , 5% CO2 , balance N2 ) during plethysmography. However, OVX rats at both 2 and 12 weeks demonstrated a persistent loss of AIH-induced pLTF relative to control animals (P < 0.01), suggesting that other sources of oestradiol synthesis were insufficient to restore pLTF. These data are consistent with our previous work indicating that oestradiol plays a key role in expression of AIH-induced respiratory neuroplasticity.
Collapse
Affiliation(s)
- Rebecca Barok
- Rehabilitation Science Graduate Program, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Jessica M L Grittner
- Rehabilitation Science Graduate Program, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Brendan J Dougherty
- Divisions of Physical Therapy and Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
8
|
Rashidy-Pour A, Derafshpour L, Vafaei AA, Bandegi AR, Kashefi A, Sameni HR, Jashire-Nezhad N, Saboory E, Panahi Y. Effects of treadmill exercise and sex hormones on learning, memory and hippocampal brain-derived neurotrophic factor levels in transient congenital hypothyroid rats. Behav Pharmacol 2021; 31:641-651. [PMID: 32826427 DOI: 10.1097/fbp.0000000000000572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Transient thyroid function abnormalities at birth exhibit intellectual developmental and cognitive disorders in adulthood. Given the well-known effects of physical activity and sex hormones on cognitive functions and brain-derived neurotrophic factor (BDNF), the present study examined the effects of treadmill exercise, sex hormones, and the combined treatment on learning and memory and hippocampal BDNF levels in transient congenital hypothyroid rats. To induce hypothyroidism, 6-propyl-2-thiouracil was added to the drinking water from the 6th day of gestation to the 21st postnatal day (PND). From PNDs 28 to 47, female and male pup rats received 17β-estradiol and testosterone, respectively, and about 30 min later, they were forced to run on the treadmill for 30 min once a day. On PNDs 48-55, spatial learning and memory of all rats tested in the water maze, which followed by measurement of BDNF in the hippocampus. Results showed that developmental hypothyroidism induced significant deficits in spatial learning and memory and hippocampal BDNF in both male and female rats. In both male and female hypothyroid rats, exercise and exercise plus sex hormones, but not sex hormones alone alleviated learning and memory deficits and all treatments (exercise, sex hormones, and the combined treatment) increased hippocampal BDNF. These disconnects in the effects of exercise, sex hormones and the combined treatment on behavioral and neurochemical outcomes suggest that a neurochemical mechanism other than hippocampal BDNF might contribute in the ameliorating effects of exercise on learning and memory deficits induced by developmental thyroid hormone insufficiency.
Collapse
Affiliation(s)
- Ali Rashidy-Pour
- Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan
| | - Leila Derafshpour
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia
| | - Abbas Ali Vafaei
- Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan
| | - Ahmad Reza Bandegi
- Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan.,Department of Biochemistry, Faculty of Medicine, Semnan University of Medical Sciences
| | - Adel Kashefi
- Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan
| | - Hamid Reza Sameni
- Research Center of Nervous System Stem Cell, Semnan University of Medical sciences, Semnan
| | - Nahid Jashire-Nezhad
- Research Center of Nervous System Stem Cell, Semnan University of Medical sciences, Semnan
| | - Ehsan Saboory
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia
| | - Yosef Panahi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| |
Collapse
|
9
|
Astrocyte-Derived Estrogen Regulates Reactive Astrogliosis and is Neuroprotective following Ischemic Brain Injury. J Neurosci 2020; 40:9751-9771. [PMID: 33158962 DOI: 10.1523/jneurosci.0888-20.2020] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 12/14/2022] Open
Abstract
Expression of the 17β-estradiol (E2) synthesis enzyme aromatase is highly upregulated in astrocytes following brain injury. However, the precise role of astrocyte-derived E2 in the injured brain remains unclear. In the current study, we generated a glial fibrillary acidic protein (GFAP) promoter-driven aromatase knock-out (GFAP-ARO-KO) mouse model to deplete astrocyte-derived E2 in the brain and determine its roles after global cerebral ischemia (GCI) in male and female mice. GFAP-ARO-KO mice were viable and fertile, with normal gross brain structure, normal morphology, intensity and distribution of astrocytes, normal aromatase expression in neurons, and normal cognitive function basally. In contrast, after GCI, GFAP-ARO-KO mice: (1) lacked the normal elevation of astrocyte aromatase and hippocampal E2 levels; (2) had significantly attenuated reactive astrogliosis; and (3) displayed enhanced neuronal damage, microglia activation, and cognitive deficits. RNA-sequencing (RNA-seq) analysis revealed that the ischemic GFAP-ARO-KO mouse hippocampus failed to upregulate the "A2" panel of reactive astrocyte genes. In addition, the JAK-STAT3 pathway, which is critical for the induction of reactive astrogliosis, was significantly downregulated in the GFAP-ARO-KO hippocampus following GCI. Finally, exogenous E2 administration fully rescued the compromised JAK-STAT3 pathway and reactive astrogliosis, and reversed the enhanced neuronal damage and microglial activation in the GFAP-ARO-KO mice after GCI, suggesting that the defects in the KO mice are because of a loss of E2 rather than an increase in precursor androgens. In conclusion, the current study provides novel genetic evidence for a beneficial role of astrocyte-derived E2 in reactive astrogliosis, microglial activation, and neuroprotection following an ischemic injury to the brain.SIGNIFICANCE STATEMENT Following cerebral ischemia, reactive astrocytes express the enzyme aromatase and produce 17β-estradiol (E2), although the precise role of astrocyte-derived E2 is poorly understood. In this study, we generated a glial fibrillary acidic protein (GFAP) promoter-driven aromatase knock-out (GFAP-ARO-KO) mouse to deplete astrocyte-derived E2 and elucidate its roles after global cerebral ischemia (GCI). The GFAP-ARO-KO mice exhibited significantly attenuated reactive astrogliosis, as well as enhanced microglial activation, neuronal damage, and cognitive dysfunction after GCI. Transcriptome analysis further revealed that astrocyte-derived E2 was critical for the induction of the JAK-STAT3 signaling pathway, as well as the A2 reactive astrocyte phenotype after ischemia. Collectively, these findings indicate that astrocyte-derived E2 has a key role in the regulation of reactive astrogliosis, microglial activation, and neuroprotection after cerebral ischemia.
Collapse
|
10
|
Notaras M, van den Buuse M. Neurobiology of BDNF in fear memory, sensitivity to stress, and stress-related disorders. Mol Psychiatry 2020; 25:2251-2274. [PMID: 31900428 DOI: 10.1038/s41380-019-0639-2] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/01/2019] [Accepted: 12/12/2019] [Indexed: 01/17/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is widely accepted for its involvement in resilience and antidepressant drug action, is a common genetic locus of risk for mental illnesses, and remains one of the most prominently studied molecules within psychiatry. Stress, which arguably remains the "lowest common denominator" risk factor for several mental illnesses, targets BDNF in disease-implicated brain regions and circuits. Altered stress-related responses have also been observed in animal models of BDNF deficiency in vivo, and BDNF is a common downstream intermediary for environmental factors that potentiate anxiety- and depressive-like behavior. However, BDNF's broad functionality has manifested a heterogeneous literature; likely reflecting that BDNF plays a hitherto under-recognized multifactorial role as both a regulator and target of stress hormone signaling within the brain. The role of BDNF in vulnerability to stress and stress-related disorders, such as posttraumatic stress disorder (PTSD), is a prominent example where inconsistent effects have emerged across numerous models, labs, and disciplines. In the current review we provide a contemporary update on the neurobiology of BDNF including new data from the behavioral neuroscience and neuropsychiatry literature on fear memory consolidation and extinction, stress, and PTSD. First we present an overview of recent advances in knowledge on the role of BDNF within the fear circuitry, as well as address mounting evidence whereby stress hormones interact with endogenous BDNF-TrkB signaling to alter brain homeostasis. Glucocorticoid signaling also acutely recruits BDNF to enhance the expression of fear memory. We then include observations that the functional common BDNF Val66Met polymorphism modulates stress susceptibility as well as stress-related and stress-inducible neuropsychiatric endophenotypes in both man and mouse. We conclude by proposing a BDNF stress-sensitivity hypothesis, which posits that disruption of endogenous BDNF activity by common factors (such as the BDNF Val66Met variant) potentiates sensitivity to stress and, by extension, vulnerability to stress-inducible illnesses. Thus, BDNF may induce plasticity to deleteriously promote the encoding of fear and trauma but, conversely, also enable adaptive plasticity during extinction learning to suppress PTSD-like fear responses. Ergo regulators of BDNF availability, such as the Val66Met polymorphism, may orchestrate sensitivity to stress, trauma, and risk of stress-induced disorders such as PTSD. Given an increasing interest in personalized psychiatry and clinically complex cases, this model provides a framework from which to experimentally disentangle the causal actions of BDNF in stress responses, which likely interact to potentiate, produce, and impair treatment of, stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia. .,College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia. .,Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
11
|
Xiao-Yao-San Formula Improves Cognitive Ability by Protecting the Hippocampal Neurons in Ovariectomized Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4156145. [PMID: 32655660 PMCID: PMC7321526 DOI: 10.1155/2020/4156145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 12/27/2022]
Abstract
Xiao-Yao-San (XYS) decoction is a traditional Chinese medicine formula. This study aimed to investigate the effect of XYS on cognitive abilities and its underlying mechanism in ovariectomized rats. Female Sprague-Dawley rats were ovariectomized and treated with XYS (3 g/kg or 9 g/kg) by gavage, with subcutaneous injection of 17-β estradiol (E2, 2 μg/kg) as a positive drug control and gavage of 1 ml saline (0.9%) as a placebo control. After 6 weeks of treatment, rats were examined using the Morris water maze test. The estradiol level in the serum and hippocampus was measured by ELISA. Golgi staining was performed to observe neuronal morphology in the hippocampus. Apoptosis of hippocampal cells was observed by TUNEL staining. The protein content of N-methyl-D-aspartate receptor (NMDAR) 2A and 2B in the hippocampal CA1 region was determined by Western blot and immunohistochemistry. Expression of estrogen receptor (ER) and PI3K signaling was detected by Western blot. Compared with the sham group, both learning and memory were impaired in ovariectomized rats. Rats treated with E2 or high-dose XYS showed better learning and memory compared with the saline-treated rats. High-dose XYS significantly reduced escape latency in the spatial acquisition trial; meanwhile, the cross times and duration in the probe quadrant were increased in the spatial probe trial. High-dose XYS promoted the de novo synthesis of E2 content in the hippocampus but had no significant effect on the serum E2 level. Golgi staining indicated that high-dose XYS could increase the branch number and density of dendritic spines in the hippocampal CA1 area. TUNEL staining showed that high-dose XYS alleviated ovariectomy-induced neuronal apoptosis. The expression level of NMDAR2A and NMDAR2B in hippocampal CA1 was upregulated by XYS treatment. The beneficial effect of XYS was through activating ERα-PI3K signaling. In conclusion, high-dose XYS treatment can improve the cognitive abilities of ovariectomized rats by protecting the hippocampal neurons and restoring the hippocampal E2 level.
Collapse
|
12
|
Loss of Estrogen Efficacy Against Hippocampus Damage in Long-Term OVX Mice Is Related to the Reduction of Hippocampus Local Estrogen Production and Estrogen Receptor Degradation. Mol Neurobiol 2020; 57:3540-3551. [PMID: 32542593 DOI: 10.1007/s12035-020-01960-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/28/2020] [Indexed: 10/24/2022]
Abstract
Postmenopausal women experience a higher risk for neurodegenerative diseases, including cognitive impairment and ischemic stroke. Many preclinical studies have indicated that estrogen replacement therapy (ERT) may provide protective effects against these neurological diseases. However, the results of Women's Health Initiative (WHI) studies have led to the proposal of "critical period hypothesis," which states that there is a precise window of opportunity for administering beneficial hormone therapy following menopause. However, the underlying molecular mechanisms require further characterization. Here, we explored the effects of ERT on cognition decline and global cerebral ischemia (GCI)-induced hippocampal neuronal damage in mice that had experienced both short-term (ovariectomized (OVX) 1 week) and long-term (OVX 10 weeks) estrogen deprivation. We also further explored the concentration of 17β-estradiol (E2) in the circulation and hippocampus and the expression of aromatase and estrogen receptors (ERα, ERα-Ser118, and ERβ). We found that the neuroprotective effectiveness of ERT against hippocampus damage exhibited in OVX1w mice was totally absent in OVX10w mice. Interestingly, the concentration of hippocampal E2 was irreversibly reduced in OVX10w mice, which was related to the decrease of aromatase expression in the hippocampus. In addition, long-term estrogen deprivation (LTED) led to a decrease in estrogen receptor proteins in the hippocampus. Thus, we concluded that the loss of ERT neuroprotection against hippocampus injury in LTED mice was related to the reduction in hippocampus E2 production and estrogen receptor degradation. These results provide several intervention targets to restore the effectiveness of ERT neuroprotection in elderly post-menopausal women.
Collapse
|
13
|
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: 14] [Impact Index Per Article: 3.5] [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
|
14
|
Tantipongpiradet A, Monthakantirat O, Vipatpakpaiboon O, Khampukdee C, Umehara K, Noguchi H, Fujiwara H, Matsumoto K, Sekeroglu N, Kijjoa A, Chulikhit Y. Effects of Puerarin on the Ovariectomy-Induced Depressive-Like Behavior in ICR Mice and Its Possible Mechanism of Action. Molecules 2019; 24:molecules24244569. [PMID: 31847138 PMCID: PMC6943479 DOI: 10.3390/molecules24244569] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/05/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022] Open
Abstract
Daily treatment of ovariectomized (OVX) ICR mice with puerarin, a glycosyl isoflavone isolated from the root bark of Pueraria candollei var. mirifica, and 17β-estradiol attenuated ovariectomy-induced depression-like behavior, as indicated by a decrease in immobility times in the tail suspension test (TST) and the forced swimming test (FST), an increase in the uterine weight and volume, a decrease in serum corticosterone levels, and dose-dependently normalized the downregulated transcription of the brain-derived neurotrophic factor (BDNF) and estrogen receptor (Erβ and Erα) mRNAs. Like 17β-estradiol, puerarin also inhibited ovariectomy-induced suppression of neurogenesis in the dentate gyrus of the hippocampus (increased the number of doublecortin (DCX)-immunosuppressive cells). These results suggest that puerarin exerts antidepressant-like effects in OVX animals, possibly by attenuating the OVX-induced hyperactivation of the HPA axis and/or normalizing the downregulated transcription of BDNF and ER mRNA in the brain.
Collapse
Affiliation(s)
- Ariyawan Tantipongpiradet
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (A.T.); (O.M.); (O.V.); (C.K.)
| | - Orawan Monthakantirat
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (A.T.); (O.M.); (O.V.); (C.K.)
| | - Onchuma Vipatpakpaiboon
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (A.T.); (O.M.); (O.V.); (C.K.)
| | - Charinya Khampukdee
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (A.T.); (O.M.); (O.V.); (C.K.)
| | - Kaoru Umehara
- Department of Pharmacognosy, School of Pharmaceutical Sciences, University of Shizuoka, Yada 52-1, Shizuoka-shi, Shizuoka 422-8526, Japan; (K.U.); (H.N.)
| | - Hiroshi Noguchi
- Department of Pharmacognosy, School of Pharmaceutical Sciences, University of Shizuoka, Yada 52-1, Shizuoka-shi, Shizuoka 422-8526, Japan; (K.U.); (H.N.)
| | - Hironori Fujiwara
- Division of Medicinal Pharmacology, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (H.F.); (K.M.)
| | - Kinzo Matsumoto
- Division of Medicinal Pharmacology, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (H.F.); (K.M.)
| | - Nazim Sekeroglu
- Department of Horticulture, Faculty of Agriculture, Killis 7 Aralik University, Killis 79000, Turkey;
| | - Anake Kijjoa
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar and CIIMAR, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- Correspondence: (A.K.); (Y.C.); Tel.: +351-220428331 (A.K.)
| | - Yaowared Chulikhit
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (A.T.); (O.M.); (O.V.); (C.K.)
- Correspondence: (A.K.); (Y.C.); Tel.: +351-220428331 (A.K.)
| |
Collapse
|
15
|
Kandasamy M, Radhakrishnan RK, Poornimai Abirami GP, Roshan SA, Yesudhas A, Balamuthu K, Prahalathan C, Shanmugaapriya S, Moorthy A, Essa MM, Anusuyadevi M. Possible Existence of the Hypothalamic-Pituitary-Hippocampal (HPH) Axis: A Reciprocal Relationship Between Hippocampal Specific Neuroestradiol Synthesis and Neuroblastosis in Ageing Brains with Special Reference to Menopause and Neurocognitive Disorders. Neurochem Res 2019; 44:1781-1795. [PMID: 31254250 DOI: 10.1007/s11064-019-02833-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/13/2019] [Accepted: 06/19/2019] [Indexed: 12/18/2022]
|
16
|
Detection of estradiol in rat brain tissues: Contribution of local versus systemic production. Psychoneuroendocrinology 2019; 102:84-94. [PMID: 30529907 DOI: 10.1016/j.psyneuen.2018.11.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 11/22/2018] [Accepted: 11/25/2018] [Indexed: 11/24/2022]
Abstract
Estrogens play important roles in regulating brain development, brain function, and behavior. Many studies have evaluated these effects using ovariectomized (OVX) rats or mice with different doses of estrogen replacement, assuming that estradiol levels in all regions of the brain are the same as levels achieved in the serum. It is well known, however, that the brain contains all the enzymes necessary to produce estrogens, and that estrogen levels in the brain are determined by both systemic and local production and are region-specific. The present study conducted a detailed analysis of the relationship between systemic levels of 17-β-estradiol (E2) achieved by estrogen replacement and levels achieved in specific regions of the brain. Levels of E2 were measured in both brain and serum in OVX rats treated with different doses of estradiol benzoate (EB) using a novel and recently validated UPLC-MS/MS method. Results confirmed significantly higher levels of E2 in the brain than in serum in brain regions known to contain aromatase (ARO) activity, both in OVX controls and in rats treated with physiological doses of EB. Additional studies compared the level of E2 and testosterone (T) in the brain and serum between testosterone propionate (TP) treated OVX and male. This demonstrated higher levels of E2 in certain brain regions of males than in TP treated OVX females even though T levels in the brain and serum were similar between the two groups. Studies also demonstrated that the differences between serum and brain levels of E2 can be eliminated by letrozole (ARO inhibitor) treatment, which indicates that the differences are due to local ARO activity. Collectively the results provide a detailed analysis of brain region-specific E2 concentrations in OVX, E2-, and T-treated rats and demonstrate the degree to which these concentrations are ARO-dependent.
Collapse
|
17
|
Derafshpour L, Saboory E, Vafaei AA, Rashidy-Pour A, Roshan-Milani S, Rasmi Y, Panahi Y, Sameni H. Interactive Effects of Exercise, Sex Hormones, and Transient Congenital Hypothyroidism on Long-Term Potentiation in Hippocampal Slices of Rat Offspring. Basic Clin Neurosci 2019; 10:119-135. [PMID: 31031899 PMCID: PMC6484195 DOI: 10.32598/bcn.9.10.170] [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: 11/13/2017] [Revised: 12/25/2017] [Accepted: 04/30/2018] [Indexed: 11/20/2022] Open
Abstract
Introduction: The long-term adverse effects of transient thyroid function abnormalities at birth on intellectual development are proven. The effect of exercise increases in the presence of sex hormones. The current study aimed at investigating the possibility that a combination of sex hormones and exercise has synergistic effects on neural plasticity in Transient Congenital Hypothyroidism (TCH) rats. Methods: To induce hypothyroidism in the mothers, Propylthiouracil (PTU) was added to drinking water (100 mg/L) on the 6th day of gestation and continued until the 21st Postnatal Day. From Postnatal Day (PND) 28 to 47, the female and male pups received 17β-estradiol and testosterone, respectively. The mild treadmill exercise began 30 minutes after the sex hormones or vehicle administration. On PND 48, electrophysiological experiments were performed on brain slices. Results: Increase of Long-Term Potentiation (LTP) was observed in sedentary-non-hormone female rats of TCH group, compared with that of the control. The exercise enhanced LTP in control rats, but the hormones showed no significant effect. The effect of exercise and sex hormone was not significant in the TCH group. The combination of exercise and testosterone enhanced LTP in TCH male rats, while the combination of exercise and estradiol or each of them individually did not produce such an effect on LTP in TCH female rats. Conclusion: The study findings showed an increase in excitatory transmission despite the returning of thyroid hormone levels to normal range in TCH female rats. Also a combination treatment including exercise and testosterone enhanced LTP in male rats of TCH group, which was a gender-specific event.
Collapse
Affiliation(s)
- Leila Derafshpour
- Laboratory of Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.,Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Ehsan Saboory
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Ali Vafaei
- Laboratory of Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Laboratory of Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Shiva Roshan-Milani
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Panahi
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Hamidreza Sameni
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
18
|
Baud O, Berkane N. Hormonal Changes Associated With Intra-Uterine Growth Restriction: Impact on the Developing Brain and Future Neurodevelopment. Front Endocrinol (Lausanne) 2019; 10:179. [PMID: 30972026 PMCID: PMC6443724 DOI: 10.3389/fendo.2019.00179] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
The environment in which a fetus develops is not only important for its growth and maturation but also for its long-term postnatal health and neurodevelopment. Several hormones including glucocorticosteroids, estrogens and progesterone, insulin growth factor and thyroid hormones, carefully regulate the growth of the fetus and its metabolism during pregnancy by controlling the supply of nutrients crossing the placenta. In addition to fetal synthesis, hormones regulating fetal growth are also expressed and regulated in the placenta, and they play a key role in the vulnerability of the developing brain and its maturation. This review summarizes the current understanding and evidence regarding the involvement of hormonal dysregulation associated with intra-uterine growth restriction and its consequences on brain development.
Collapse
Affiliation(s)
- Olivier Baud
- Division of Neonatology and Pediatric Intensive Care, Department of Women-Children-Teenagers, University Hospitals Geneva, Geneva, Switzerland
- Inserm U1141, Sorbonne, Paris Diderot University, Paris, France
- *Correspondence: Olivier Baud
| | - Nadia Berkane
- Division of Obstetrics and Gynecology, Department of Women-Children-Teenagers, University Hospitals Geneva, Geneva, Switzerland
| |
Collapse
|
19
|
Rune GM. Einfluss von Sexualhormonen auf die Kognition. GYNAKOLOGISCHE ENDOKRINOLOGIE 2018. [DOI: 10.1007/s10304-018-0213-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
Kraus K, Kleene R, Henis M, Braren I, Kataria H, Sharaf A, Loers G, Schachner M, Lutz D. A Fragment of Adhesion Molecule L1 Binds to Nuclear Receptors to Regulate Synaptic Plasticity and Motor Coordination. Mol Neurobiol 2018; 55:7164-7178. [PMID: 29383692 DOI: 10.1007/s12035-018-0901-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/10/2018] [Indexed: 02/05/2023]
Abstract
Proteolytic cleavage of the neuronal isoform of the murine cell adhesion molecule L1, triggered by stimulation of the cognate L1-dependent signaling pathways, results in the generation and nuclear import of an L1 fragment that contains the intracellular domain, the transmembrane domain, and part of the extracellular domain. Here, we show that the LXXLL and FXXLF motifs in the extracellular and transmembrane domain of this L1 fragment mediate the interaction with the nuclear estrogen receptors α (ERα) and β (ERβ), peroxisome proliferator-activated receptor γ (PPARγ), and retinoid X receptor β (RXRβ). Mutations of the LXXLL motif in the transmembrane domain and of the FXXLF motif in the extracellular domain disturb the interaction of the L1 fragment with these nuclear receptors and, when introduced by viral transduction into mouse embryos in utero, result in impaired motor coordination, learning and memory, as well as synaptic connectivity in the cerebellum, in adulthood. These impairments are similar to those observed in the L1-deficient mouse. Our findings suggest that the interplay of nuclear L1 and distinct nuclear receptors is associated with synaptic contact formation and plasticity.
Collapse
Affiliation(s)
- Kristina Kraus
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ralf Kleene
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Melad Henis
- Institut für Strukturelle Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Ingke Braren
- Vector Core Unit, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Hardeep Kataria
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ahmed Sharaf
- Institut für Strukturelle Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Gabriele Loers
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China.
| | - David Lutz
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
- Institut für Strukturelle Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
21
|
Sex Differences in Neuropathology and Cognitive Behavior in APP/PS1/tau Triple-Transgenic Mouse Model of Alzheimer's Disease. Neurosci Bull 2018; 34:736-746. [PMID: 30099679 DOI: 10.1007/s12264-018-0268-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 06/16/2018] [Indexed: 10/28/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia among the elderly, characterized by amyloid plaques, neurofibrillary tangles, and neuroinflammation in the brain, as well as impaired cognitive behaviors. A sex difference in the prevalence of AD has been noted, while sex differences in the cerebral pathology and relevant molecular mechanisms are not well clarified. In the present study, we systematically investigated the sex differences in pathological characteristics and cognitive behavior in 12-month-old male and female APP/PS1/tau triple-transgenic AD mice (3×Tg-AD mice) and examined the molecular mechanisms. We found that female 3×Tg-AD mice displayed more prominent amyloid plaques, neurofibrillary tangles, neuroinflammation, and spatial cognitive deficits than male 3×Tg-AD mice. Furthermore, the expression levels of hippocampal protein kinase A-cAMP response element-binding protein (PKA-CREB) and p38-mitogen-activated protein kinases (MAPK) also showed sex difference in the AD mice, with a significant increase in the levels of p-PKA/p-CREB and a decrease in the p-p38 in female, but not male, 3×Tg-AD mice. We suggest that an estrogen deficiency-induced PKA-CREB-MAPK signaling disorder in 12-month-old female 3×Tg-AD mice might be involved in the serious pathological and cognitive damage in these mice. Therefore, sex differences should be taken into account in investigating AD biomarkers and related target molecules, and estrogen supplementation or PKA-CREB-MAPK stabilization could be beneficial in relieving the pathological damage in AD and improving the cognitive behavior of reproductively-senescent females.
Collapse
|
22
|
Mantor D, Pratchayasakul W, Minta W, Sutham W, Palee S, Sripetchwandee J, Kerdphoo S, Jaiwongkum T, Sriwichaiin S, Krintratun W, Chattipakorn N, Chattipakorn SC. Both oophorectomy and obesity impaired solely hippocampal-dependent memory via increased hippocampal dysfunction. Exp Gerontol 2018; 108:149-158. [PMID: 29678475 DOI: 10.1016/j.exger.2018.04.010] [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] [Received: 12/16/2017] [Revised: 03/18/2018] [Accepted: 04/16/2018] [Indexed: 02/06/2023]
Abstract
Our previous study demonstrated that obesity aggravated peripheral insulin resistance and brain dysfunction in the ovariectomized condition. Conversely, the effect of obesity followed by oophorectomy on brain oxidative stress, brain apoptosis, synaptic function and cognitive function, particularly in hippocampal-dependent and hippocampal-independent memory, has not been investigated. Our hypothesis was that oophorectomy aggravated metabolic impairment, brain dysfunction and cognitive impairment in obese rats. Thirty-two female rats were fed with either a normal diet (ND, n = 16) or a high-fat diet (HFD, n = 16) for a total of 20 weeks. At week 13, rats in each group were subdivided into sham and ovariectomized subgroups (n = 8/subgroup). At week 20, all rats were tested for hippocampal-dependent and hippocampal-independent memory by using Morris water maze test (MWM) and Novel objective recognition (NOR) tests, respectively. We found that the obese-insulin resistant condition occurred in sham-HFD-fed rats (HFS), ovariectomized-ND-fed rats (NDO), and ovariectomized-HFD-fed rats (HFO). Increased hippocampal oxidative stress level, increased hippocampal apoptosis, increased hippocampal synaptic dysfunction, decreased hippocampal estrogen level and impaired hippocampal-dependent memory were observed in HFS, NDO, and HFO rats. However, the hippocampal-independent memory, cortical estrogen levels, cortical ROS production, and cortical apoptosis showed no significant difference between groups. These findings suggested that oophorectomy and obesity exclusively impaired hippocampal-dependent memory, possibly via increased hippocampal dysfunction. Nonetheless, oophorectomy did not aggravate these deleterious effects under conditions of obesity.
Collapse
Affiliation(s)
- Duangkamol Mantor
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wasana Pratchayasakul
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wanitchaya Minta
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wissuta Sutham
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siripong Palee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jirapas Sripetchwandee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thidarat Jaiwongkum
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirawit Sriwichaiin
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Warunsorn Krintratun
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
23
|
Munk AJL, Zoeller AC, Hennig J. Fluctuations of estradiol during women's menstrual cycle: Influences on reactivity towards erotic stimuli in the late positive potential. Psychoneuroendocrinology 2018. [PMID: 29518692 DOI: 10.1016/j.psyneuen.2018.02.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND While several studies examined the reactivity towards negative emotional stimuli across women's menstrual cycle, only few investigated responses to positive emotional cues in association with sexual hormones on a neural level. Therefore, the aim of the current EEG-experiment was to study the differential reactivity towards positive (erotic) words during the menstrual cycle (i.e. with fluctuations in the steroids estradiol and progesterone) in the late positive potential (LPP). Regarding reactivity towards erotic stimuli, the LPP is seen as the most relevant ERP-component, as more positive amplitudes in the LPP reflect larger incentive salience and higher arousal. The LPP towards erotic words was expected to be more pronounced during fertile phases of the menstrual cycle (around ovulation). Furthermore, associations with hormonal concentrations of estradiol and progesterone were investigated. METHOD 19 young, free cycling women were tested in an Erotic Stroop paradigm during the follicular phase, ovulation, and the luteal phase in a balanced cross-over design, while electroencephalogram (EEG) was recorded. RESULTS LPPs in reaction to erotic compared to neutral words were larger in every phase. During the follicular phase and ovulation, higher estradiol-concentrations were associated with more positive LPP-amplitudes towards erotic- than to neutral words. No effects of progesterone, as well as no effects of cycle phase, were evident. Results are being discussed regarding implications for further research.
Collapse
Affiliation(s)
- Aisha J L Munk
- Department of Differential and Biological Psychology, University of Giessen, Germany.
| | - Aaron C Zoeller
- Department of General Psychology, University of Giessen, Germany
| | - Juergen Hennig
- Department of Differential and Biological Psychology, University of Giessen, Germany
| |
Collapse
|
24
|
Akinola OB, Gabriel MO. Neuroanatomical and molecular correlates of cognitive and behavioural outcomes in hypogonadal males. Metab Brain Dis 2018; 33:491-505. [PMID: 29230619 DOI: 10.1007/s11011-017-0163-5] [Citation(s) in RCA: 5] [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: 07/12/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022]
Abstract
Robust epidemiological, clinical and laboratory evidence supports emerging roles for the sex steroids in such domains as neurodevelopment, behaviour, learning and cognition. Regions of the mammalian brain that are involved in cognitive development and memory do not only express the classical nuclear androgen receptor, but also the non-genomic membrane receptor, which is a G protein-coupled receptor that mediates some rapid effects of the androgens on neurogenesis and synaptic plasticity. Under physiological conditions, hippocampal neurons do express the enzyme aromatase, and therefore actively aromatize testosterone to oestradiol. Although glial expression of the aromatase enzyme is minimal, increased expression following injury suggests a role for sex steroids in neuroprotection. It is therefore plausible to deduce that low levels of circulating androgens in males would perturb neuronal functions in relation to cognition and memory, as well as neural repair following injury. The present review is an overview of some roles of the sex steroids on cognitive function in males, and the neuroanatomical and molecular underpinnings of some behavioural and cognitive deficits characteristic of such genetic disorders noted for low androgen levels, including Klinefelter syndrome, Bardet-Biedl syndrome, Kallman syndrome and Prader-Willi syndrome. Recent literature in relation to some behavioural and cognitive changes secondary to surgical and pharmacological castration are also appraised.
Collapse
Affiliation(s)
- O B Akinola
- Division of Endocrinology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.
| | - M O Gabriel
- Division of Endocrinology, Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
25
|
Chamniansawat S, Sawatdiyaphanon C. Age-Related Memory Impairment Associated With Decreased Endogenous Estradiol in the Hippocampus of Female Rats. Int J Toxicol 2018; 37:207-215. [PMID: 29554823 DOI: 10.1177/1091581818761653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It is widely known that not only the gonadal estradiol (E2) but also hippocampal E2 plays an essential role in memory process. However, the role of hippocampal E2-enhanced memory mechanism during aging is largely unknown. The aim of the present study was to investigate the effect of age on E2 concentration, the expression level of its receptors, and key steroidogenic enzymes in hippocampus. We also investigated the effect of microglia activation on E2 synthesis in hippocampal neurons. The results showed that serum E2 was higher in 19-month-old (aged) rats, which exhibited spatial memory decline in the Morris water maze (MWM) test when compared to the younger rats. Hence, serum E2 may not be associated with the reduced spatial memory performance in aging. In contrast, the level of E2 and the expressions of its receptors were significantly decreased in hippocampus of aged female rat compared to younger females. Furthermore, the expressions of key hippocampal steroidogenic enzymes, steroidogenic acute regulatory protein (StAR), and cytochrome P450 (P450) also significantly decreased with age, which resulted in lower hippocampal E2 levels. In addition, we found that the microglia of aged brain highly expressed interleukin 6 (IL-6), which directly inhibited E2 synthesis in hippocampal neurons via suppression of P450 synthesis. Taken together, we summarized that the microglia-derived IL-6 inhibited hippocampal E2 synthesis in aged rats which, in turn, contributed to the deficit of spatial memory performance.
Collapse
Affiliation(s)
- Siriporn Chamniansawat
- 1 Faculty of Allied Health Sciences, Department of Biomedical Sciences, Burapha University, Chonburi, Thailand
| | - Chattraporn Sawatdiyaphanon
- 1 Faculty of Allied Health Sciences, Department of Biomedical Sciences, Burapha University, Chonburi, Thailand
| |
Collapse
|
26
|
Hwang DS, Kim N, Choi JG, Kim HG, Kim H, Oh MS. Dangguijakyak-san ameliorates memory deficits in ovariectomized mice by upregulating hippocampal estrogen synthesis. Altern Ther Health Med 2017; 17:501. [PMID: 29178947 PMCID: PMC5702078 DOI: 10.1186/s12906-017-2015-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 11/17/2017] [Indexed: 01/20/2023]
Abstract
Background Dangguijakyak-san (DJS) is an herbal formulation that has been clinically applicable for treating postmenopausal symptoms and neurological disorders. It is reported that hippocampal estrogen attenuates memory impairment via neuroprotection and synaptogenesis. However, the effect of DJS on hippocampal estrogen synthesis remains unknown. In this study, we explored the effect of DJS and its neuroprotective mechanism against memory impairment in ovariectomized (OVX) mice, with respect to hippocampal estrogen stimulation. Methods Cell cultures were prepared from the hippocampi of 18-day-old embryos from timed pregnant Sprague–Dawley rats. The hippocampi were dissected, collected, dissociated, and plated in 60-mm dishes. The cells were treated with DJS for 48 h and the supernatant was collected to determine estrogen levels. Female ICR mice (8-weeks-old) were housed for 1 week and ovariectomy was performed to remove the influence of ovary-synthesized estrogens. Following a 2-week post-surgical recovery period, the mice were administrated with DJS (50 and 100 mg/kg/day, p.o.) or 17β-estradiol (200 μg/kg/day, i.p.) once daily for 21 days. Hippocampal and serum estrogen levels were determined using enzyme-linked immunosorbent assay kit. Memory behavioral tests, western blot, and immunohistochemical analyses were performed to evaluate the neuroprotective effects of DJS in this model. Results DJS treatment promoted estrogen synthesis in primary hippocampal cells and the hippocampus of OVX mice, resulting in the amelioration of OVX-induced memory impairment. Hippocampal estrogen stimulated by DJS treatment contributed to the activation of cAMP response element-binding protein and synaptic protein in OVX mice. Conclusion DJS may attenuate memory deficits in postmenopausal women via hippocampal estrogen synthesis. Electronic supplementary material The online version of this article (10.1186/s12906-017-2015-6) contains supplementary material, which is available to authorized users.
Collapse
|
27
|
The effect of sex and menstrual phase on memory formation during a nap. Neurobiol Learn Mem 2017; 145:119-128. [DOI: 10.1016/j.nlm.2017.09.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 01/20/2023]
|
28
|
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: 56] [Impact Index Per Article: 7.0] [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
|
29
|
Circulating Estradiol Regulates Brain-Derived Estradiol via Actions at GnRH Receptors to Impact Memory in Ovariectomized Rats. eNeuro 2016; 3:eN-NWR-0321-16. [PMID: 28032117 PMCID: PMC5172373 DOI: 10.1523/eneuro.0321-16.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 11/14/2016] [Indexed: 01/09/2023] Open
Abstract
Systemic estradiol treatment enhances hippocampus-dependent memory in ovariectomized rats. Although these enhancements are traditionally thought to be due to circulating estradiol, recent data suggest these changes are brought on by hippocampus-derived estradiol, the synthesis of which depends on gonadotropin-releasing hormone (GnRH) activity. The goal of the current work is to test the hypothesis that peripheral estradiol affects hippocampus-dependent memory through brain-derived estradiol regulated via hippocampal GnRH receptor activity. In the first experiment, intracerebroventricular infusion of letrozole, which prevents the synthesis of estradiol, blocked the ability of peripheral estradiol administration in ovariectomized rats to enhance hippocampus-dependent memory in a radial-maze task. In the second experiment, hippocampal infusion of antide, a long-lasting GnRH receptor antagonist, blocked the ability of peripheral estradiol administration in ovariectomized rats to enhance hippocampus-dependent memory. In the third experiment, hippocampal infusion of GnRH enhanced hippocampus-dependent memory, the effects of which were blocked by letrozole infusion. Results indicate that peripheral estradiol-induced enhancement of cognition is mediated by brain-derived estradiol via hippocampal GnRH receptor activity.
Collapse
|
30
|
Ruiz-Palmero I, Ortiz-Rodriguez A, Melcangi RC, Caruso D, Garcia-Segura LM, Rune GM, Arevalo MA. Oestradiol synthesized by female neurons generates sex differences in neuritogenesis. Sci Rep 2016; 6:31891. [PMID: 27553191 PMCID: PMC4995407 DOI: 10.1038/srep31891] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 07/29/2016] [Indexed: 12/04/2022] Open
Abstract
Testosterone produced by the foetal testis is converted by male neurons to oestradiol, which masculinizes neuronal morphology. Female neurons are known to synthesize oestradiol in absence of exogenous testosterone. However, the role of neuronal oestradiol on the differentiation of foetal female neurons is unknown. Here we show that, due to endogenous neuronal oestradiol synthesis, female hippocampal neurons have higher expression of the neuritogenic protein Neurogenin 3 and enhanced neuritogenesis than males. Exogenous application of testosterone or its metabolite dihydrotestosterone increases Neurogenin 3 expression and promotes neuritogenesis in males, but reduces these parameters in females. Together our data indicate that gonadal-independent oestradiol synthesis by female neurons participates in the generation of sex differences in hippocampal neuronal development.
Collapse
Affiliation(s)
- Isabel Ruiz-Palmero
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - Ana Ortiz-Rodriguez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Via G. Balzaretti 9, 20133 Milan, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Via G. Balzaretti 9, 20133 Milan, Italy
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Maria-Angeles Arevalo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
| |
Collapse
|
31
|
London SE. Influences of non-canonical neurosteroid signaling on developing neural circuits. Curr Opin Neurobiol 2016; 40:103-110. [PMID: 27429051 DOI: 10.1016/j.conb.2016.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/21/2016] [Accepted: 06/22/2016] [Indexed: 12/31/2022]
Abstract
Developing neural circuits are especially susceptible to environmental perturbation. Endocrine signaling systems such as steroids provide a mechanism to encode physiological changes and integrate function across various biological systems including the brain. 'Neurosteroids' are synthesized and act within the brain across development. There is a long history of steroids sculpting developing neural circuits; more recently, evidence has demonstrated how neurosteroids influence the early potential for neural circuits to organize and transmit precise information via non-canonical receptor types.
Collapse
Affiliation(s)
- Sarah E London
- University of Chicago, Psychology, 940 E 57th Street, 125C BPSB, Chicago, IL 60637, United States.
| |
Collapse
|
32
|
Fester L, Brandt N, Windhorst S, Pröls F, Bläute C, Rune GM. Control of aromatase in hippocampal neurons. J Steroid Biochem Mol Biol 2016; 160:9-14. [PMID: 26472556 DOI: 10.1016/j.jsbmb.2015.10.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 10/01/2015] [Accepted: 10/08/2015] [Indexed: 10/22/2022]
Abstract
Our knowledge on estradiol-induced modulation of synaptic function in the hippocampus is widely based on results following the application of the steroid hormone to either cell cultures, or after the treatment of gonadectomized animals, thus ignoring local neuronal estrogen synthesis. We and others, however, have shown that hippocampus-derived estradiol also controls synaptic plasticity in the hippocampus. Estradiol synthesis in the hippocampus is regulated by several mechanisms, which are reviewed in this report. The regulation of the activity of aromatase, the final enzyme of estrogen biosynthesis, by Ca(2+) transients, is of particular interest. Aromatase becomes inactivated as soon as it is phosphorylated by Ca(2+)-dependent kinases upon calcium release from internal stores. Accordingly, thapsigargin dephosphorylates aromatase and stimulates estradiol synthesis by depletion of internal Ca(2+) stores. Vice versa, letrozole, an aromatase inhibitor, phosphorylates aromatase and reduces estradiol synthesis. Treatment of the cultures with 17β-estradiol results in phosphorylation of the enzyme and increased aromatase protein expression, which suggests that estradiol synthesis in hippocampal neurons is regulated in an autocrine manner.
Collapse
Affiliation(s)
- Lars Fester
- University Medical Center Hamburg Eppendorf, Institute of Neuroanatomy, Martinistr. 52, 20246 Hamburg, Germany
| | - Nicola Brandt
- University Medical Center Hamburg Eppendorf, Institute of Neuroanatomy, Martinistr. 52, 20246 Hamburg, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, Martinistr. 52, 20246 Hamburg, Germany
| | - Felicitas Pröls
- University Medical Center Hamburg Eppendorf, Institute of Neuroanatomy, Martinistr. 52, 20246 Hamburg, Germany
| | - Corinna Bläute
- University Medical Center Hamburg Eppendorf, Institute of Neuroanatomy, Martinistr. 52, 20246 Hamburg, Germany
| | - Gabriele M Rune
- University Medical Center Hamburg Eppendorf, Institute of Neuroanatomy, Martinistr. 52, 20246 Hamburg, Germany.
| |
Collapse
|
33
|
Qiu L, Zhao Y, Guo Q, Zhang Y, He L, Li W, Zhang J. Dose-dependent regulation of steroid receptor coactivator-1 and steroid receptors by testosterone propionate in the hippocampus of adult male mice. J Steroid Biochem Mol Biol 2016; 156:23-31. [PMID: 26607693 DOI: 10.1016/j.jsbmb.2015.11.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 01/08/2023]
Abstract
Androgens have been proposed to play important roles in the regulation of hippocampus function either directly, through the androgen receptor (AR), or indirectly, through estrogen receptors (ERs), after aromatization into estradiol. Steroid receptor coactivator-1 (SRC-1) is present in the hippocampus of several species, and its expression is regulated by development and aging, as well as by orchidectomy and aromatase inhibitor letrozole administration, while ovariectomy only transiently downregulated hippocampal SRC-1. However, whether the expression of hippocampal SRC-1 can be directly regulated by testosterone, the principal male sex hormone, remains unclear. In the present study, we investigated the expression of hippocampal SRC-1 after orchidectomy and testosterone treatment using immunohistochemistry and Western blot analysis. We found that while hippocampal SRC-1 was significantly downregulated by orchidectomy (ORX), its expression was rescued by treatment with testosterone in a dose-dependent manner. Furthermore, we noticed that the decreased expression of hippocampal AR, ERs and the synaptic proteins GluR-1 and PSD-95 induced by ORX was also rescued by testosterone treatment in a dose-dependent manner. However, we found that hippocampal membrane estrogen receptor GPR30 and dendritic spine marker spinophilin were not altered by ORX or testosterone treatment. Together, the above results provided the first direct evidence for the androgenic regulation on hippocampal SRC-1, indicating that SRC-1 may be a direct target of androgenic regulation on the hippocampus. Furthermore, because AR and ERs can be differentially regulated by testosterone, and the transcriptional activity requires the involvement of local SRC-1, and considering the complicated regulatory pathway of each individual receptor, the converged hub regulator SRC-1 of these nuclear receptor networks is worthy of further investigation.
Collapse
Affiliation(s)
- Linli Qiu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China; Department of Filed Nursing, School of Nursing, Third Military Medical University, Chongqing 400038, China
| | - Yangang Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Qiang Guo
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Yuanyuan Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Li He
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China; Department of Filed Nursing, School of Nursing, Third Military Medical University, Chongqing 400038, China
| | - Wei Li
- Department of Filed Nursing, School of Nursing, Third Military Medical University, Chongqing 400038, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
34
|
Ferris JK, Tse MT, Hamson DK, Taves MD, Ma C, McGuire N, Arckens L, Bentley GE, Galea LAM, Floresco SB, Soma KK. Neuronal Gonadotrophin-Releasing Hormone (GnRH) and Astrocytic Gonadotrophin Inhibitory Hormone (GnIH) Immunoreactivity in the Adult Rat Hippocampus. J Neuroendocrinol 2015; 27:772-86. [PMID: 26258544 DOI: 10.1111/jne.12307] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/03/2015] [Accepted: 08/04/2015] [Indexed: 01/17/2023]
Abstract
Gonadotrophin-releasing hormone (GnRH) and gonadotrophin inhibitory hormone (GnIH) are neuropeptides secreted by the hypothalamus that regulate reproduction. GnRH receptors are not only present in the anterior pituitary, but also are abundantly expressed in the hippocampus of rats, suggesting that GnRH regulates hippocampal function. GnIH inhibits pituitary gonadotrophin secretion and is also expressed in the hippocampus of a songbird; its role outside of the reproductive axis is not well established. In the present study, we employed immunohistochemistry to examine three forms of GnRH [mammalian GnRH-I (mGnRH-I), chicken GnRH-II (cGnRH-II) and lamprey GnRH-III (lGnRH-III)] and GnIH in the adult rat hippocampus. No mGnRH-I and cGnRH-II+ cell bodies were present in the hippocampus. Sparse mGnRH-I and cGnRH-II+ fibres were present within the CA1 and CA3 fields of the hippocampus, along the hippocampal fissure, and within the hilus of the dentate gyrus. No lGnRH-III was present in the rodent hippocampus. GnIH-immunoreactivity was present in the hippocampus in cell bodies that resembled astrocytes. Males had more GnIH+ cells in the hilus of the dentate gyrus than females. To confirm the GnIH+ cell body phenotype, we performed double-label immunofluorescence against GnIH, glial fibrillary acidic protein (GFAP) and NeuN. Immunofluorescence revealed that all GnIH+ cell bodies in the hippocampus also contained GFAP, a marker of astrocytes. Taken together, these data suggest that GnRH does not reach GnRH receptors in the rat hippocampus primarily via synaptic release. By contrast, GnIH might be synthesised locally in the rat hippocampus by astrocytes. These data shed light on the sites of action and possible functions of GnRH and GnIH outside of the hypothalamic-pituitary-gonadal axis.
Collapse
Affiliation(s)
- J K Ferris
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - M T Tse
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - D K Hamson
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - M D Taves
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - C Ma
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - N McGuire
- Department of Integrative Biology, University of California-Berkeley, Berkeley, CA, USA
| | - L Arckens
- Laboratory of Neuroplasticity and Neuroproteomics, University of Leuven, Leuven, Belgium
| | - G E Bentley
- Department of Integrative Biology, University of California-Berkeley, Berkeley, CA, USA
| | - L A M Galea
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - S B Floresco
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - K K Soma
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
35
|
Duarte-Guterman P, Yagi S, Chow C, Galea LAM. Hippocampal learning, memory, and neurogenesis: Effects of sex and estrogens across the lifespan in adults. Horm Behav 2015; 74:37-52. [PMID: 26122299 DOI: 10.1016/j.yhbeh.2015.05.024] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/29/2015] [Accepted: 05/26/2015] [Indexed: 01/12/2023]
Abstract
This article is part of a Special Issue "Estradiol and Cognition". There are sex differences in hippocampus-dependent cognition and neurogenesis suggesting that sex hormones are involved. Estrogens modulate certain forms of spatial and contextual memory and neurogenesis in the adult female rodent, and to a lesser extent male, hippocampus. This review focuses on the effects of sex and estrogens on hippocampal learning, memory, and neurogenesis in the young and aged adult rodent. We discuss how factors such as the type of estrogen, duration and dose of treatment, timing of treatment, and type of memory influence the effects of estrogens on cognition and neurogenesis. We also address how reproductive experience (pregnancy and mothering) and aging interact with estrogens to modulate hippocampal cognition and neurogenesis in females. Given the evidence that adult hippocampal neurogenesis plays a role in long-term spatial memory and pattern separation, we also discuss the functional implications of regulating neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Paula Duarte-Guterman
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Shunya Yagi
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Carmen Chow
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Liisa A M Galea
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
36
|
|
37
|
Schmidt PJ, Dor RB, Martinez PE, Guerrieri GM, Harsh VL, Thompson K, Koziol DE, Nieman LK, Rubinow DR. Effects of Estradiol Withdrawal on Mood in Women With Past Perimenopausal Depression: A Randomized Clinical Trial. JAMA Psychiatry 2015; 72:714-26. [PMID: 26018333 PMCID: PMC6391160 DOI: 10.1001/jamapsychiatry.2015.0111] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
IMPORTANCE Perimenopause is accompanied by an increased risk of new and recurrent depression. The coincidence of declining ovarian function with the onset of depression led to the inference that "withdrawal" from physiologic estradiol levels underpinned depression in perimenopause. To our knowledge, this is the first controlled systematic study to directly test the estrogen withdrawal theory of perimenopausal depression (PMD). OBJECTIVE To examine the role of estradiol withdrawal in PMD. DESIGN, SETTING, AND PARTICIPANTS Initial open-label treatment with estradiol followed by randomized, double-blind, placebo-controlled, parallel-design evaluation of continued estradiol treatment was evaluated at an outpatient research facility at the National Institutes of Health Clinical Center. An intent-to-treat analysis was performed between October 2003 and July 2012. Participants included asymptomatic postmenopausal women with past PMD responsive to hormone therapy (n = 26) and asymptomatic postmenopausal women with no history of depression (n = 30) matched for age, body mass index, and reproductive status who served as controls. Data were analyzed between November 2012 and October 2013 by repeated-measures analysis of variance. INTERVENTIONS After 3 weeks of open-label administration of transdermal estradiol (100 µg/d), participants were randomized to a parallel design to receive either estradiol (100 µg/d; 27 participants) or matched placebo skin patches (29 participants) for 3 additional weeks under double-blind conditions. MAIN OUTCOMES AND MEASURES Center for Epidemiologic Studies-Depression Scale and 17-item Hamilton Depression Rating Scale (completed by raters blind to diagnosis and randomization status), self-administered visual analog symptom ratings, and blood hormone levels obtained at weekly clinic visits. RESULTS None of the women reported depressive symptoms during open-label use of estradiol. Women with past PMD who were crossed over from estradiol to placebo experienced a significant increase in depression symptom severity demonstrated using the Center for Epidemiologic Studies-Depression Scale and 17-item Hamilton Depression Rating Scale, with mean (SD) scores increasing from estradiol (ie, 2.4 [2.0] and 3.0 [2.5]) to placebo (8.8 [4.9] and 6.6 [4.5], respectively [P = .0004 for both]). Women with past PMD who continued estradiol therapy and all women in the control group remained asymptomatic. Women in both groups had similar hot-flush severity and plasma estradiol levels during use of placebo. CONCLUSIONS AND RELEVANCE In women with past PMD that was previously responsive to hormone therapy, the recurrence of depressive symptoms during blinded hormone withdrawal suggests that normal changes in ovarian estradiol secretion can trigger an abnormal behavioral state in these susceptible women. Women with a history of PMD should be alert to the risk of recurrent depression when discontinuing hormone therapy. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00060736.
Collapse
Affiliation(s)
- Peter J. Schmidt
- Section on Behavioral Endocrinology, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Rivka Ben Dor
- Section on Behavioral Endocrinology, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Pedro E. Martinez
- Section on Behavioral Endocrinology, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Gioia M. Guerrieri
- Section on Behavioral Endocrinology, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Veronica L. Harsh
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville
| | - Karla Thompson
- Section on Behavioral Endocrinology, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Deloris E. Koziol
- Biostatistics and Clinical Epidemiology Service, Clinical Center, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Lynnette K. Nieman
- Intramural Research Program on Reproductive and Adult Endocrinology, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - David R. Rubinow
- Department of Psychiatry, University of North Carolina, Chapel Hill
| |
Collapse
|
38
|
Bethea CL, Phu K, Kim A, Reddy AP. Androgen metabolites impact CSF amines and axonal serotonin via MAO-A and -B in male macaques. Neuroscience 2015; 301:576-89. [PMID: 26086546 DOI: 10.1016/j.neuroscience.2015.06.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/02/2015] [Accepted: 06/11/2015] [Indexed: 01/11/2023]
Abstract
A number of studies have shown that mutations or deletions of the monoamine oxidase-A (MAO-A) gene cause elevated CNS serotonin and elevated impulsive aggression in humans and animal models. In addition, low cerebrospinal fluid (CSF) 5-hydroxyindole acetic acid (5HIAA) has been documented in a limited number of violent criminal populations and in macaques that exhibit impulsive aggression. To reconcile these different analyses, we hypothesized that CSF 5HIAA reflected degradation of serotonin by the activity of MAO-A; and that low MAO-A activity would result in lower CSF 5HIAA, but overall higher serotonin in the CNS. To test this hypothesis, male Japanese macaques (Macaca fuscata) were castrated, rested for 5-7months, and then treated for 3months with [1] placebo, [2] testosterone (T), [3] dihydrotestosterone (DHT; non-aromatizable androgen) and 1,4,6-androstatriene-3,17-dione (ATD) (steroidal aromatase inhibitor), or [4] flutamide (FLUT; androgen antagonist) and ATD (n=5/group). These treatments enable isolation of androgen and estrogen activities. In the dorsal raphe, MAO-A and MAO-B expressions were determined with in situ hybridization (ISH) and protein expression of aromatase was determined with immunohistochemistry (IHC). CSF concentrations of 5HIAA, 3-methoxy-4-hydroxyphenylglycol (MHPG), and homovanillic acid (HVA) were determined with liquid chromatography/mass spectrometry (LC/MS). From the same animals, previously published data on serotonin axon density were used as a proxy for CNS serotonin. Aromatase conversion of T to estrogen (E) suppressed MAO-A (positive pixel area, p=0.0045), but androgens increased MAO-B (positive pixel area, p=0.014). CSF 5HIAA was suppressed by conversion of T to E (Cohen's d=0.6). CSF 5HIAA was positively correlated with MAO-A-positive pixel area (r(2)=0.78). CSF 5HIAA was inversely correlated with serotonin axon-positive pixel area (r(2)=0.69). In summary, CSF 5HIAA reflects MAO-A activity rather than global serotonin. Low CSF 5HIAA may, in this paradigm, reflect higher serotonin activity. Androgens lower MAO-A activity via metabolism to E, thus elevating CNS serotonin and decreasing CSF 5HIAA. Since androgens increase certain types of aggression, these data are consistent with studies demonstrating that lower MAO-A activity is associated with elevated serotonin and increased aggression.
Collapse
Affiliation(s)
- C L Bethea
- Division of Reproductive and Development Science, Oregon National Primate Research Center, Beaverton, OR 97006, United States; Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, United States; Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97201, United States.
| | - K Phu
- Division of Reproductive and Development Science, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| | - A Kim
- Division of Reproductive and Development Science, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| | - A P Reddy
- Division of Reproductive and Development Science, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| |
Collapse
|
39
|
Cornil CA, Ball GF, Balthazart J. The dual action of estrogen hypothesis. Trends Neurosci 2015; 38:408-16. [PMID: 26089224 DOI: 10.1016/j.tins.2015.05.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/18/2015] [Accepted: 05/24/2015] [Indexed: 11/25/2022]
Abstract
Estradiol (E2) can act in the brain in a relatively fast manner (i.e., seconds to minutes) usually through signaling initiated at the cell membrane. Brain-derived E2 has thus been considered as another type of neurotransmitter. Recent work found that behaviors indicative of male sexual motivation are activated by estrogenic metabolites of testosterone (T) in a fast manner, while sexual performance (copulatory behavior per se) is regulated by brain E2 in a slower manner via nucleus-initiated actions. This functional division between these two types of action appears to generalize to other behavioral systems regulated by E2. We propose the dual action of estrogen hypothesis to explain this functional distinction between these two different modes of action.
Collapse
Affiliation(s)
- Charlotte A Cornil
- GIGA Neurosciences, University of Liege, Quartier Hôpital, 15 Avenue Hippocrate, 4000 Liège, Belgium
| | - Gregory F Ball
- Department of Psychology, 2141 Tydings Hall, University of Maryland, College Park, MD 20742-7201, USA
| | - Jacques Balthazart
- GIGA Neurosciences, University of Liege, Quartier Hôpital, 15 Avenue Hippocrate, 4000 Liège, Belgium.
| |
Collapse
|
40
|
Heimovics SA, Trainor BC, Soma KK. Rapid Effects of Estradiol on Aggression in Birds and Mice: The Fast and the Furious. Integr Comp Biol 2015; 55:281-93. [PMID: 25980562 DOI: 10.1093/icb/icv048] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Across invertebrates and vertebrates, steroids are potent signaling molecules that affect nearly every cell in the organism, including cells of the nervous system. Historically, researchers have focused on the genomic (or "nuclear-initiated") effects of steroids. However, all classes of steroids also have rapid non-genomic (or "membrane-initiated") effects, although there is far less basic knowledge of these non-genomic effects. In particular, steroids synthesized in the brain ("neurosteroids") have genomic and non-genomic effects on behavior. Here, we review evidence that estradiol has rapid effects on aggression, an important social behavior, and on intracellular signaling cascades in relevant regions of the brain. In particular, we focus on studies of song sparrows (Melospiza melodia) and Peromyscus mice, in which estradiol has rapid behavioral effects under short photoperiods only. Furthermore, in captive Peromyscus, estrogenic compounds (THF-diols) in corncob bedding profoundly alter the rapid effects of estradiol. Environmental factors in the laboratory, such as photoperiod, diet, and bedding, are critical variables to consider in experimental design. These studies are consistent with the hypothesis that locally-produced steroids are more likely than systemic steroids to act via non-genomic mechanisms. Furthermore, these studies illustrate the dynamic balance between genomic and non-genomic signaling for estradiol, which is likely to be relevant for other steroids, behaviors, and species.
Collapse
Affiliation(s)
- Sarah A Heimovics
- *Department of Biology, University of St Thomas, St Paul, MN 55105, USA;
| | - Brian C Trainor
- Department of Psychology, University of California-Davis, Davis, CA 95616, USA
| | - Kiran K Soma
- Departments of Psychology and Zoology, Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, V6T 1Z7, Canada
| |
Collapse
|
41
|
G-protein-coupled estrogen receptor 1 is anatomically positioned to modulate synaptic plasticity in the mouse hippocampus. J Neurosci 2015; 35:2384-97. [PMID: 25673833 DOI: 10.1523/jneurosci.1298-14.2015] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Both estrous cycle and sex affect the numbers and types of neuronal and glial profiles containing the classical estrogen receptors α and β, and synaptic levels in the rodent dorsal hippocampus. Here, we examined whether the membrane estrogen receptor, G-protein-coupled estrogen receptor 1 (GPER1), is anatomically positioned in the dorsal hippocampus of mice to regulate synaptic plasticity. By light microscopy, GPER1-immunoreactivity (IR) was most noticeable in the pyramidal cell layer and interspersed interneurons, especially those in the hilus of the dentate gyrus. Diffuse GPER1-IR was found in all lamina but was most dense in stratum lucidum of CA3. Ultrastructural analysis revealed discrete extranuclear GPER1-IR affiliated with the plasma membrane and endoplasmic reticulum of neuronal perikarya and dendritic shafts, synaptic specializations in dendritic spines, and clusters of vesicles in axon terminals. Moreover, GPER1-IR was found in unmyelinated axons and glial profiles. Overall, the types and amounts of GPER1-labeled profiles were similar between males and females; however, in females elevated estrogen levels generally increased axonal labeling. Some estradiol-induced changes observed in previous studies were replicated by the GPER agonist G1: G1 increased PSD95-IR in strata oriens, lucidum, and radiatum of CA3 in ovariectomized mice 6 h after administration. In contrast, estradiol but not G1 increased Akt phosphorylation levels. Instead, GPER1 actions in the synapse may be due to interactions with synaptic scaffolding proteins, such as SAP97. These results suggest that although estrogen's actions via GPER1 may converge on the same synaptic elements, different pathways are used to achieve these actions.
Collapse
|
42
|
Opioid receptor-dependent sex differences in synaptic plasticity in the hippocampal mossy fiber pathway of the adult rat. J Neurosci 2015; 35:1723-38. [PMID: 25632146 DOI: 10.1523/jneurosci.0820-14.2015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The mossy fiber (MF) pathway is critical to hippocampal function and influenced by gonadal hormones. Physiological data are limited, so we asked whether basal transmission and long-term potentiation (LTP) differed in slices of adult male and female rats. The results showed small sex differences in basal transmission but striking sex differences in opioid receptor sensitivity and LTP. When slices were made from females on proestrous morning, when serum levels of 17β-estradiol peak, the nonspecific opioid receptor antagonist naloxone (1 μm) enhanced MF transmission but there was no effect in males, suggesting preferential opioid receptor-dependent inhibition in females when 17β-estradiol levels are elevated. The μ-opioid receptor (MOR) antagonist Cys2,Tyr3,Orn5,Pen7-amide (CTOP; 300 nm) had a similar effect but the δ-opioid receptor (DOR) antagonist naltrindole (NTI; 1 μm) did not, implicating MORs in female MF transmission. The GABAB receptor antagonist saclofen (200 μm) occluded effects of CTOP but the GABAA receptor antagonist bicuculline (10 μm) did not. For LTP, a low-frequency (LF) protocol was used because higher frequencies elicited hyperexcitability in females. Proestrous females exhibited LF-LTP but males did not, suggesting a lower threshold for synaptic plasticity when 17β-estradiol is elevated. NTI blocked LF-LTP in proestrous females, but CTOP did not. Electron microscopy revealed more DOR-labeled spines of pyramidal cells in proestrous females than males. Therefore, we suggest that increased postsynaptic DORs mediate LF-LTP in proestrous females. The results show strong MOR regulation of MF transmission only in females and identify a novel DOR-dependent form of MF LTP specific to proestrus.
Collapse
|
43
|
Wang H, Si L, Li X, Deng W, Yang H, Yang Y, Fu Y. Overexpression of estrogen receptor beta alleviates the toxic effects of beta-amyloid protein on PC12 cells via non-hormonal ligands. Neural Regen Res 2015; 7:1095-100. [PMID: 25722700 PMCID: PMC4340023 DOI: 10.3969/j.issn.1673-5374.2012.14.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Accepted: 04/23/2012] [Indexed: 01/08/2023] Open
Abstract
After binding to the estrogen receptor, estrogen can alleviate the toxic effects of beta-amyloid protein, and thereby exert a therapeutic effect on Alzheimer's disease patients. Estrogen can increase the incidence of breast carcinoma and endometrial cancer in post-menopausal women, so it is not suitable for clinical treatment of Alzheimer's disease. There is recent evidence that the estrogen receptor can exert its neuroprotective effects without estrogen dependence. Real-time quantitative PCR and flow cytometry results showed that, compared with non-transfected PC12 cells, adenovirus-mediated estrogen receptor β gene-transfected PC12 cells exhibited lower expression of tumor necrosis factor α and interleukin 1β under stimulation with beta-amyloid protein and stronger protection from apoptosis. The Akt-specific inhibitor Abi-2 decreased the anti-inflammatory and anti-apoptotic effects of estrogen receptor β gene-transfection. These findings suggest that overexpression of estrogen receptor β can alleviate the toxic effect of beta-amyloid protein on PC12 cells, without estrogen dependence. The Akt pathway is one of the potential means for the anti-inflammatory and anti-apoptotic effects of the estrogen receptor.
Collapse
Affiliation(s)
- Hui Wang
- Department of Gynaecology and Obstetrics, First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Lihui Si
- Department of Gynaecology and Obstetrics, Second Hospital of Jilin University, Changchun 130044, Jilin Province, China
| | - Xiaoxi Li
- Department of Gynaecology, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Weiguo Deng
- Department of Children's Heatlh, School of Public Health, Jilin Univeristy, Changchun 130021, China
| | - Haimiao Yang
- Department of Scientific Research, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130021, China
| | - Yuyan Yang
- Department of Gynaecology and Obstetrics, Minzu Hospital of Yitong County, Yitong 130700, Jilin Province, China
| | - Yan Fu
- Department of Gynaecology and Obstetrics, First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
44
|
Fokidis HB, Adomat HH, Kharmate G, Hosseini-Beheshti E, Guns ES, Soma KK. Regulation of local steroidogenesis in the brain and in prostate cancer: lessons learned from interdisciplinary collaboration. Front Neuroendocrinol 2015; 36:108-29. [PMID: 25223867 DOI: 10.1016/j.yfrne.2014.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 11/16/2022]
Abstract
Sex steroids play critical roles in the regulation of the brain and many other organs. Traditionally, researchers have focused on sex steroid signaling that involves travel from the gonads via the circulation to intracellular receptors in target tissues. This classic concept has been challenged, however, by the growing number of cases in which steroids are synthesized locally and act locally within diverse tissues. For example, the brain and prostate carcinoma were previously considered targets of gonadal sex steroids, but under certain circumstances, these tissues can upregulate their steroidogenic potential, particularly when circulating sex steroid concentrations are low. We review some of the similarities and differences between local sex steroid synthesis in the brain and prostate cancer. We also share five lessons that we have learned during the course of our interdisciplinary collaboration, which brought together neuroendocrinologists and cancer biologists. These lessons have important implications for future research in both fields.
Collapse
Affiliation(s)
- H Bobby Fokidis
- Department of Biology, Rollins College, Winter Park, FL 37289, USA; Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada.
| | - Hans H Adomat
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | | | | | - Emma S Guns
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada; Department of Urological Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kiran K Soma
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Brain Research Centre, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
45
|
Ransome MI. Could androgens maintain specific domains of mental health in aging men by preserving hippocampal neurogenesis? Neural Regen Res 2014; 7:2227-39. [PMID: 25538744 PMCID: PMC4268723 DOI: 10.3969/j.issn.1673-5374.2012.028.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 07/10/2012] [Indexed: 12/18/2022] Open
Abstract
Interest surrounds the role of sex-hormones in regulating brain function outside of reproductive behaviour. Declining androgen production in aging males has been associated with cognitive impairment, depression and increased risk of developing Alzheimer's disease. Indication for testosterone replacement therapy is based on biochemically determined low circulating testosterone combined with manifest symptoms. However, which aspects of age-related cognitive decline are attributable to low circulating testosterone remain ambiguous. Studies examining cognition in aging men receiving testosterone replacement therapy have yielded equivocal results. The exact role of testosterone in maintaining cognitive function and the underlying neural mechanisms are largely unknown, though it would appear to be domain specific. Clarity in this area will provide clinical direction toward addressing an increasing healthcare burden of mental health decline coincident with increasing longevity. The premise that androgens contribute to maintaining aspects of mental health in aging men by preserving hippocampal neurogenesis will be used as a forum in this review to discuss current knowledge and the need for further studies to better define testosterone replacement strategies for aging male health.
Collapse
Affiliation(s)
- Mark I Ransome
- Florey Neurosciences Institute, Melbourne Brain Centre, the University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
46
|
Chamniansawat S, Chongthammakun S. Inhibition of hippocampal estrogen synthesis by reactive microglia leads to down-regulation of synaptic protein expression. Neurotoxicology 2014; 46:25-34. [PMID: 25447322 DOI: 10.1016/j.neuro.2014.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 11/12/2014] [Accepted: 11/12/2014] [Indexed: 12/31/2022]
Abstract
Activation of microglia may facilitate age-related impairment in cognitive functions including hippocampal-dependent memory. Considerable evidence indicates that hippocampal-derived estrogen improves hippocampal-dependent learning and memory. We hypothesize that activated microglia may inhibit de novo hippocampal estrogen synthesis and in turn suppress hippocampal synaptic protein expression. The present study aimed to elucidate the role of lipopolysaccharide (LPS)-activated microglial HAPI cells on estrogen synthesis and expression of synaptic proteins using H19-7 hippocampal neurons with a neuron-microglia co-culture system. LPS induced expression of the microglial activation markers major histocompatibility complex II (MHC II), CD11b, and ionized calcium-binding adapter molecule 1 (Iba1). Prolonged LPS exposure also enhanced the secretion of interleukin (IL)-6 and nitric oxide (NO) from microglial HAPI cells. Exposure to either LPS-activated microglia or IL-6, significantly suppressed the expression of synaptic proteins and the secretion of de novo hippocampal estrogen in H19-7 hippocampal neurons. In addition, LPS-activated microglia also decreased the expression of estrogen receptors (ERα and ERβ) in H19-7 hippocampal neurons. Our findings demonstrate a potential mechanism of microglia activation underlying the reduction in estrogen-mediated signaling on synaptic proteins in hippocampal neurons, which may be involved in hippocampal-dependent memory formation.
Collapse
Affiliation(s)
- Siriporn Chamniansawat
- Division of Anatomy, Department of Biomedical Sciences, Faculty of Allied Health Sciences, Burapha University, 169 Long-Hard Bangsaen Road, SaenSook Sub-district, Mueang District, Chonburi 20131, Thailand.
| | - Sukumal Chongthammakun
- Department of Anatomy and Center for Neuroscience, Faculty of Science, Mahidol University, 272 Rama VI Rd., Ratchathewi District, Bangkok 10400, Thailand
| |
Collapse
|
47
|
Ikeda T, Makino Y, Yamada MK. 17α-estradiol is generated locally in the male rat brain and can regulate GAD65 expression and anxiety. Neuropharmacology 2014; 90:9-14. [PMID: 25446575 DOI: 10.1016/j.neuropharm.2014.10.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 10/18/2014] [Indexed: 12/23/2022]
Abstract
Increasing evidence suggests that 17β-estradiol, a sex hormone, is synthesized by neurons. In addition, 17α-estradiol, the stereoisomer of 17β-estradiol, is reported to be the dominant form in the male mouse brain. However, probably because the method to detect these isomers requires unusual and precise experimental design, the presence of this endogenous 17α-estradiol has not been reported subsequently and the actual role is therefore not well elucidated. We first quantified the estradiol level in hippocampal extracts using gas chromatography/mass spectrometry. As a result, 17α-estradiol was found in all of the male rats tested, while that of 17β-estradiol was detected only in a certain subset. The estrogen-biosynthesis inhibitor letrozole decreased the expression of the major presynaptic GABA synthesizing enzyme GAD65 in cultured neurons and the effect was abrogated by exogenously supplied 17α-estradiol. Next, injection of the inhibitor into the brain reduced the 17α-estradiol level, indicating its biogenesis in the brain. Under the same conditions, immuno-staining of GAD65 was also decreased. Furthermore, the inhibitor treatment increased anxiety index of rats in the open field and this was ameliorated by the addition of 17α-estradiol. We showed that 17α-estradiol was generated in the brain and acted as a regulator of inhibitory neurotransmission as well as behavior. These results may have implications for a variety of diseases, such as the menopausal depression and Alzheimer's disease that have been reported to be related to estrogen levels.
Collapse
Affiliation(s)
- Takamitsu Ikeda
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yukiko Makino
- Laboratory of Chemistry and Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Maki K Yamada
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; PRESTO, The Japan Science and Technology Agency, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
48
|
Ghorbanpoor S, Garcia-Segura LM, Haeri-Rohani A, Khodagholi F, Jorjani M. Aromatase inhibition exacerbates pain and reactive gliosis in the dorsal horn of the spinal cord of female rats caused by spinothalamic tract injury. Endocrinology 2014; 155:4341-55. [PMID: 25105782 DOI: 10.1210/en.2014-1158] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Central pain syndrome is characterized by severe and excruciating pain resulting from a lesion in the central nervous system. Previous studies have shown that estradiol decreases pain and that inhibitors of the enzyme aromatase, which synthesizes estradiol from aromatizable androgens, increases pain sensitivity. In this study we have assessed whether aromatase expression in the dorsal horns of the spinal cord is altered in a rat model of central pain syndrome, induced by the unilateral electrolytic lesion of the spinothalamic tract. Protein and mRNA levels of aromatase, as well as the protein and mRNA levels of estrogen receptors α and β, were increased in the dorsal horn of female rats after spinothalamic tract injury, suggesting that the injury increased estradiol synthesis and signaling in the dorsal horn. To determine whether the increased aromatase expression in this pain model may participate in the control of pain, mechanical allodynia thresholds were determined in both hind paws after the intrathecal administration of letrozole, an aromatase inhibitor. Aromatase inhibition enhanced mechanical allodynia in both hind paws. Because estradiol is known to regulate gliosis we assessed whether the spinothalamic tract injury and aromatase inhibition regulated gliosis in the dorsal horn. The proportion of microglia with a reactive phenotype and the number of glial fibrillary acidic protein-immunoreactive astrocytes were increased by the injury in the dorsal horn. Aromatase inhibition enhanced the effect of the injury on gliosis. Furthermore, a significant a positive correlation of mechanical allodynia and gliosis in the dorsal horn was detected. These findings suggest that aromatase is up-regulated in the dorsal horn in a model of central pain syndrome and that aromatase activity in the spinal cord reduces mechanical allodynia by controlling reactive gliosis in the dorsal horn.
Collapse
Affiliation(s)
- Samar Ghorbanpoor
- Department of Cell and Molecular Biology (S.G.), Department of Animal Biology (A.H.-R.), School of Biology, College of Science (S.G.), University of Tehran, Tehran, Iran; Consejo Superior de Investigaciones Científicas (L.M.G.-S.), Instituto Cajal, E-28002 Madrid, Spain; Neurobiology Research Center (F.K., M.J.), Shahid Beheshti University of Medical Sciences, Tehran, Iran; and Department of Pharmacology, Faculty of Medicine (M.J.), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | | |
Collapse
|
49
|
Scharfman HE, MacLusky NJ. Sex differences in the neurobiology of epilepsy: a preclinical perspective. Neurobiol Dis 2014; 72 Pt B:180-92. [PMID: 25058745 DOI: 10.1016/j.nbd.2014.07.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/08/2014] [Accepted: 07/11/2014] [Indexed: 10/25/2022] Open
Abstract
When all of the epilepsies are considered, sex differences are not always clear, despite the fact that many sex differences are known in the normal brain. Sex differences in epilepsy in laboratory animals are also unclear, although robust effects of sex on seizures have been reported, and numerous effects of gonadal steroids have been shown throughout the rodent brain. Here we discuss several reasons why sex differences in seizure susceptibility are unclear or are difficult to study. Examples of robust sex differences in laboratory rats, such as the relative resistance of adult female rats to the chemoconvulsant pilocarpine compared to males, are described. We also describe a novel method that has shed light on sex differences in neuropathology, which is a relatively new technique that will potentially contribute to sex differences research in the future. The assay we highlight uses the neuronal nuclear antigen NeuN to probe sex differences in adult male and female rats and mice. In females, weak NeuN expression defines a sex difference that previous neuropathological studies have not described. We also show that in adult rats, social isolation stress can obscure the normal effects of 17β-estradiol to increase excitability in area CA3 of the hippocampus. These data underscore the importance of controlling behavioral stress in studies of seizure susceptibility in rodents and suggest that behavioral stress may be one factor that has led to inconsistencies in outcomes of sex differences research. These and other issues have made it difficult to translate our increasing knowledge about the effects of gonadal hormones on the brain to improved treatment for men and women with epilepsy.
Collapse
Affiliation(s)
- Helen E Scharfman
- Department of Child & Adolescent Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA; Department of Physiology & Neuroscience, New York University Langone Medical Center, New York, NY 10016, USA; Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA.
| | - Neil J MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
50
|
Expression of estrogen receptors α and β in the trigeminal mesencephalic nucleus of adult women and men. Ann Anat 2014; 196:416-22. [PMID: 25060183 DOI: 10.1016/j.aanat.2014.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 06/03/2014] [Accepted: 06/03/2014] [Indexed: 01/01/2023]
Abstract
Temporomandibular disorders are more prevalent in women than in men and phases of pain relate to the estrous cycle. Several studies described the location of estrogen receptors (ER) in the temporomandibular joint (TMJ), the masseteric muscles and cartilage, but it was unknown whether they are also expressed within the pseudounipolar neurons of the trigeminal mesencephalic nucleus, which receives direct sensory inputs from these structures. Therefore, we studied expression of ERα and ERβ protein in the trigeminal mesencephalic nucleus of ten human brains (five female/five male). Both receptors were uniformly expressed on neurons, but not other cell types within the target structure. Thus, sensory inputs from the TMJ and adjacent structures are likely to be modulated by estrogen at the level of the first sensory neuron which may underlie the well-known correlation of pain incidence and phases of the estrous cycle.
Collapse
|