1
|
Lymer J, Bergman H, Yang S, Mallick R, Galea LAM, Choleris E, Fergusson D. The effects of estrogens on spatial learning and memory in female rodents - A systematic review and meta-analysis. Horm Behav 2024; 164:105598. [PMID: 38968677 DOI: 10.1016/j.yhbeh.2024.105598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/01/2024] [Accepted: 06/19/2024] [Indexed: 07/07/2024]
Abstract
Estrogens have inconsistent effects on learning and memory in both the clinical and preclinical literature. Preclinical literature has the advantage of investigating an array of potentially important factors contributing to the varied effects of estrogens on learning and memory, with stringently controlled studies. This study set out to identify specific factors in the animal literature that influence the effects of estrogens on cognition, for possible translation back to clinical practice. The literature was screened and studies meeting strict inclusion criteria were included in the analysis. Eligible studies included female ovariectomized rodents with an adequate vehicle for the estrogen treatment, with an outcome of spatial learning and memory in the Morris water maze. Training days of the Morris water maze were used to assess acquisition of spatial learning, and the probe trial was used to evaluate spatial memory recall. Continuous outcomes were pooled using a random effects inverse variance method and reported as standardized mean differences with 95 % confidence intervals. Subgroup analyses were developed a priori to assess important factors. The overall analysis favoured treatment for the later stages of training and for the probe trial. Factors including the type of estrogen, route, schedule of administration, age of animals, timing relative to ovariectomy, and duration of treatment were all found to be important. The subgroup analyses showed that chronic treatment with 17β-estradiol, either cyclically or continuously, to young animals improved spatial recall. These results, observed in animals, can inform and guide further clinical research on hormone replacement therapy for cognitive benefits.
Collapse
Affiliation(s)
- Jennifer Lymer
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada.
| | - Hailey Bergman
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | - Sabrina Yang
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | | | - Liisa A M Galea
- Department of Psychiatry, University of Toronto, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada.
| | - Dean Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
2
|
Wang X, Jiang Y, Feng B, Ma X, Zhang K, Yang F, Liu Z, Yang L, Yue J, Lu L, Song D, Guo Q, Qi J, Li X, Wang M, Zhang H, Huang J, Zhao M, Liu S. PJA1 mediates the effects of astrocytic GPR30 on learning and memory in female mice. J Clin Invest 2023; 133:e165812. [PMID: 37712419 PMCID: PMC10503807 DOI: 10.1172/jci165812] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 07/19/2023] [Indexed: 09/16/2023] Open
Abstract
Hormone replacement therapy (HRT) is not recommended for treating learning and memory decline in menopausal women because it exerts adverse effects by activating classic estrogen receptors ERα and ERβ. The membrane estrogen receptor G protein-coupled receptor 30 (GPR30) has been reported to be involved in memory modulation; however, the underlying mechanisms are poorly understood. Here, we found that GPR30 deletion in astrocytes, but not in neurons, impaired learning and memory in female mice. Astrocytic GPR30 depletion induced A1 phenotype transition, impairing neuronal function. Further exploration revealed that Praja1 (PJA1), a RING ubiquitin ligase, mediated the effects of astrocytic GPR30 on learning and memory by binding to Serpina3n, which is a molecular marker of neuroinflammation in astrocytes. GPR30 positively modulated PJA1 expression through the CREB signaling pathway in cultured murine and human astrocytes. Additionally, the mRNA levels of GPR30 and PJA1 were reduced in exosomes isolated from postmenopausal women while Serpina3n levels were increased in the plasma. Together, our findings suggest a key role for astrocytic GPR30 in the learning and memory abilities of female mice and identify GPR30/PJA1/Serpina3n as potential therapeutic targets for learning and memory loss in peri- and postmenopausal women.
Collapse
Affiliation(s)
| | - Yongli Jiang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Ban Feng
- Department of Pharmacology, School of Pharmacy and
| | - Xue Ma
- Department of Pharmacology, School of Pharmacy and
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy and
| | - Fan Yang
- Department of Pharmacology, School of Pharmacy and
| | - Zhenguo Liu
- Department of Pharmacy, Northwest Women’s and Children’s Hospital, Xi’an, China
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Jiao Yue
- Department of Pharmacology, School of Pharmacy and
| | - Liang Lu
- Department of Pharmacology, School of Pharmacy and
| | - Dake Song
- Department of Pharmacology, School of Pharmacy and
| | - Qingjuan Guo
- Department of Pharmacology, School of Pharmacy and
| | - Jingyu Qi
- Department of Pharmacology, School of Pharmacy and
| | - Xubo Li
- Department of Pharmacology, School of Pharmacy and
| | - Min Wang
- Department of Pharmacology, School of Pharmacy and
| | - Huinan Zhang
- Department of Health Management, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Jing Huang
- Department of Health Management, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Minggao Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Shuibing Liu
- Department of Pharmacology, School of Pharmacy and
| |
Collapse
|
3
|
Echeverria V, Echeverria F, Barreto GE, Echeverría J, Mendoza C. Estrogenic Plants: to Prevent Neurodegeneration and Memory Loss and Other Symptoms in Women After Menopause. Front Pharmacol 2021; 12:644103. [PMID: 34093183 PMCID: PMC8172769 DOI: 10.3389/fphar.2021.644103] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 04/15/2021] [Indexed: 12/11/2022] Open
Abstract
In mammals, sexual hormones such as estrogens play an essential role in maintaining brain homeostasis and function. Estrogen deficit in the brain induces many undesirable symptoms such as learning and memory impairment, sleep and mood disorders, hot flushes, and fatigue. These symptoms are frequent in women who reached menopausal age or have had ovariectomy and in men and women subjected to anti-estrogen therapy. Hormone replacement therapy alleviates menopause symptoms; however, it can increase cardiovascular and cancer diseases. In the search for therapeutic alternatives, medicinal plants and specific synthetic and natural molecules with estrogenic effects have attracted widespread attention between the public and the scientific community. Various plants have been used for centuries to alleviate menstrual and menopause symptoms, such as Cranberry, Ginger, Hops, Milk Thistle, Red clover, Salvia officinalis, Soy, Black cohosh, Turnera diffusa, Ushuva, and Vitex. This review aims to highlight current evidence about estrogenic medicinal plants and their pharmacological effects on cognitive deficits induced by estrogen deficiency during menopause and aging.
Collapse
Affiliation(s)
- Valentina Echeverria
- Facultad de Ciencias de la Salud, Universidad San Sebastian, Concepcion, Chile
- Research and Development Service, Bay Pines VA Healthcare System, Bay Pines, FL, Unites States
| | | | - George E. Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Health Research Institute, University of Limerick, Limerick, Ireland
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Cristhian Mendoza
- Facultad de Ciencias de la Salud, Universidad San Sebastian, Concepcion, Chile
| |
Collapse
|
4
|
Neuroestrogen synthesis modifies neural representations of learned song without altering vocal imitation in developing songbirds. Sci Rep 2020; 10:3602. [PMID: 32108169 PMCID: PMC7046723 DOI: 10.1038/s41598-020-60329-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/05/2020] [Indexed: 12/20/2022] Open
Abstract
Birdsong learning, like human speech, depends on the early memorization of auditory models, yet how initial auditory experiences are formed and consolidated is unclear. In songbirds, a putative cortical locus is the caudomedial nidopallium (NCM), and one mechanism to facilitate auditory consolidation is 17β-estradiol (E2), which is associated with human speech-language development, and is abundant in both NCM and human temporal cortex. Circulating and NCM E2 levels are dynamic during learning, suggesting E2’s involvement in encoding recent auditory experiences. Therefore, we tested this hypothesis in juvenile male songbirds using a comprehensive assessment of neuroanatomy, behavior, and neurophysiology. First, we found that brain aromatase expression, and thus the capacity to synthesize neuroestrogens, remains high in the auditory cortex throughout development. Further, while systemic estrogen synthesis blockade suppressed juvenile song production, neither systemic nor unilateral E2 synthesis inhibition in NCM disrupted eventual song imitation. Surprisingly, early life neuroestrogen synthesis blockade in NCM enhanced the neural representations of both the birds’ own song and the tutor song in NCM and a downstream sensorimotor region, HVC, respectively. Taken together, these findings indicate that E2 plays a multifaceted role during development, and that, contrary to prediction, tutor song memorization is unimpaired by unilateral estrogen synthesis blockade in the auditory cortex.
Collapse
|
5
|
Pickel VM, Bourie F, Chan J, Mackie K, Lane DA, Wang G. Chronic adolescent exposure to ∆9-tetrahydrocannabinol decreases NMDA current and extrasynaptic plasmalemmal density of NMDA GluN1 subunits in the prelimbic cortex of adult male mice. Neuropsychopharmacology 2020; 45:374-383. [PMID: 31323660 PMCID: PMC6901492 DOI: 10.1038/s41386-019-0466-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 07/01/2019] [Accepted: 07/12/2019] [Indexed: 12/21/2022]
Abstract
Adolescence is a vulnerable period of development when limbic connection of the prefrontal cortex (PFC) involved in emotional processing may be rendered dysfunctional by chronic exposure to delta-9-tetrahydrocannabinol (∆9-THC), the major psychoactive compound in marijuana. Cannabinoid-1 receptors (CB1Rs) largely mediate the central neural effects of ∆9-THC and endocannabinoids that regulate NMDA receptor-dependent synaptic plasticity of glutamatergic synapses in the prelimbic prefrontal cortex (PL-PFC). Thus, chronic occupancy of CB1Rs by ∆9-THC during adolescence may competitively decrease the functional expression and activity of NMDA receptors in the mature PL-PFC. We used a multidisciplinary approach to test this hypothesis in adult C57BL/6J male mice that received vehicle or ∆9-THC in escalating doses (2.5-10 mg/kg/ip) through adolescence (postnatal day 29-43). In comparison with vehicle, the mice receiving ∆9-THC showed a hyperpolarized resting membrane potential, decreased spontaneous firing rate, increased current-induced firing threshold, and decreased depolarizing response to NMDA in deep-layer PL-PFC neurons analyzed by current-clamp recordings. Electron microscopic immunolabeling in the PL-PFC of adult mice that had received Δ9-THC only during adolescence showed a significant (1) decrease in the extrasynaptic plasmalemmal density of obligatory GluN1-NMDA subunits in dendrites of all sizes and (2) a shift from cytoplasmic to plasmalemmal distribution of GluN1 in large dendrites receiving mainly inhibitory-type synapses from CB1R-labeled terminals. From these results and concomitant behavioral studies, we conclude that social dysfunctions resulting from excessive intake of ∆9-THC in the increasingly available marijuana products used by male teens may largely reflect circuit defects in PL-PFC networks communicating through endocannabinoid-regulated NMDA receptors.
Collapse
Affiliation(s)
- Virginia M Pickel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Faye Bourie
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - June Chan
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ken Mackie
- Linda and Jack Gill Center for Biomolecular Science, Dept. of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47404, USA
| | - Diane A Lane
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| |
Collapse
|
6
|
Yousuf H, Smies CW, Hafenbreidel M, Tuscher JJ, Fortress AM, Frick KM, Mueller D. Infralimbic Estradiol Enhances Neuronal Excitability and Facilitates Extinction of Cocaine Seeking in Female Rats via a BDNF/TrkB Mechanism. Front Behav Neurosci 2019; 13:168. [PMID: 31417375 PMCID: PMC6684748 DOI: 10.3389/fnbeh.2019.00168] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/11/2019] [Indexed: 11/13/2022] Open
Abstract
Women are more susceptible to developing cocaine dependence than men, but paradoxically, are more responsive to treatment. The potent estrogen, 17β-estradiol (E2), mediates these effects by augmenting cocaine seeking but also promoting extinction of cocaine seeking through E2's memory-enhancing functions. Although we have previously shown that E2 facilitates extinction, the neuroanatomical locus of action and underlying mechanisms are unknown. Here we demonstrate that E2 infused directly into the infralimbic-medial prefrontal cortex (IL-mPFC), a region critical for extinction consolidation, enhances extinction of cocaine seeking in ovariectomized (OVX) female rats. Using patch-clamp electrophysiology, we show that E2 may facilitate extinction by potentiating intrinsic excitability of IL-mPFC neurons. Because the mnemonic effects of E2 are known to be regulated by brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), we examined whether BDNF/TrkB signaling was necessary for E2-induced enhancement of excitability and extinction. We found that E2-mediated increases in excitability of IL-mPFC neurons were abolished by Trk receptor blockade. Moreover, blockade of TrkB signaling impaired E2-facilitated extinction of cocaine seeking in OVX female rats. Thus, E2 enhances IL-mPFC neuronal excitability in a TrkB-dependent manner to support extinction of cocaine seeking. Our findings suggest that pharmacological enhancement of E2 or BDNF/TrkB signaling during extinction-based therapies would improve therapeutic outcome in cocaine-addicted women.
Collapse
Affiliation(s)
- Hanna Yousuf
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Chad W Smies
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Madalyn Hafenbreidel
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Devin Mueller
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States.,Department of Biological Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
7
|
Paletta P, Sheppard PAS, Matta R, Ervin KSJ, Choleris E. Rapid effects of estrogens on short-term memory: Possible mechanisms. Horm Behav 2018; 104:88-99. [PMID: 29847771 DOI: 10.1016/j.yhbeh.2018.05.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/24/2018] [Accepted: 05/26/2018] [Indexed: 01/11/2023]
Abstract
Contribution to Special Issue on Fast effects of steroids. Estrogens affect learning and memory through rapid and delayed mechanisms. Here we review studies on rapid effects on short-term memory. Estradiol rapidly improves social and object recognition memory, spatial memory, and social learning when administered systemically. The dorsal hippocampus mediates estrogen rapid facilitation of object, social and spatial short-term memory. The medial amygdala mediates rapid facilitation of social recognition. The three estrogen receptors, α (ERα), β (ERβ) and the G-protein coupled estrogen receptor (GPER) appear to play different roles depending on the task and brain region. Both ERα and GPER agonists rapidly facilitate short-term social and object recognition and spatial memory when administered systemically or into the dorsal hippocampus and facilitate social recognition in the medial amygdala. Conversely, only GPER can facilitate social learning after systemic treatment and an ERβ agonist only rapidly improved short-term spatial memory when given systemically or into the hippocampus, but also facilitates social recognition in the medial amygdala. Investigations into the mechanisms behind estrogens' rapid effects on short term memory showed an involvement of the extracellular signal-regulated kinase (ERK) and the phosphoinositide 3-kinase (PI3K) kinase pathways. Recent evidence also showed that estrogens interact with the neuropeptide oxytocin in rapidly facilitating social recognition. Estrogens can increase the production and/or release of oxytocin and other neurotransmitters, such as dopamine and acetylcholine. Therefore, it is possible that estrogens' rapid effects on short-term memory may occur through the regulation of various neurotransmitters, although more research is need on these interactions as well as the mechanisms of estrogens' actions on short-term memory.
Collapse
Affiliation(s)
- Pietro Paletta
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Paul A S Sheppard
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Richard Matta
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Kelsy S J Ervin
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
8
|
Luine V, Serrano P, Frankfurt M. Rapid effects on memory consolidation and spine morphology by estradiol in female and male rodents. Horm Behav 2018; 104:111-118. [PMID: 29669258 PMCID: PMC6274598 DOI: 10.1016/j.yhbeh.2018.04.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/09/2018] [Accepted: 04/13/2018] [Indexed: 12/25/2022]
Abstract
Contribution to Special Issue on Fast effects of steroids. Rapid, neurosteroid-like effects of estrogens on memory consolidation during recognition memory tasks in both male and female rodents are described. We discuss how these mnemonic changes are related to rapid estrogenic effects on dendritic spine density, the distribution of spine types and the expression of PSD95 and GluA2 within spines in the hippocampus and medial prefrontal cortex, two areas critical for learning and memory. Overall, these data lead to the conclusion that estrogens are capable of exerting rapid and potent influences on memory and spine morphology in both sexes. The demonstration of estrogenic effects in males, which are used in the majority of memory studies, may provide a model for better understanding how hormone dependent changes in signaling pathways mediating memory and spinogenesis are coordinated to promote memory consolidation.
Collapse
Affiliation(s)
- Victoria Luine
- Department of Psychology, Hunter College of CUNY, New York, NY, USA.
| | - Peter Serrano
- Department of Psychology, Hunter College of CUNY, New York, NY, USA
| | - Maya Frankfurt
- Department of Science Education, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
9
|
Frick KM, Tuscher JJ, Koss WA, Kim J, Taxier LR. Estrogenic regulation of memory consolidation: A look beyond the hippocampus, ovaries, and females. Physiol Behav 2018; 187:57-66. [PMID: 28755863 PMCID: PMC5787049 DOI: 10.1016/j.physbeh.2017.07.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/14/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022]
Abstract
The potent estrogen 17β-estradiol (E2) has long been known to regulate the hippocampus and hippocampal-dependent memories in females, and research from the past decade has begun to shed light on the molecular mechanisms through which E2 mediates memory formation in females. Although E2 can also regulate hippocampal function in males, relatively little is known about how E2 influences memory formation in males, or whether sex differences in underlying mechanisms exist. This review, based on a talk given in April 2017 at the American University symposium entitled, "Sex Differences: From Neuroscience to the Clinic and Beyond", first provides an overview of the molecular mechanisms in the dorsal hippocampus through which E2 enhances memory consolidation in ovariectomized female mice. Next, newer research is described demonstrating key roles for the prefrontal cortex and de novo hippocampal E2 synthesis to the memory-enhancing effects of E2 in females. The review then discusses the effects of de novo and exogenous E2 on hippocampal memory consolidation in both sexes, and putative sex differences in the underlying molecular mechanisms through which E2 enhances memory formation. The review concludes by discussing the importance and implications of sex differences in the molecular mechanisms underlying E2-induced memory consolidation for human health.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| | - Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Wendy A Koss
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Jaekyoon Kim
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| |
Collapse
|
10
|
de Sousa LP, de Almeida RF, Ribeiro-Gomes FL, de Moura Carvalho LJ, E Souza TM, de Souza DOG, Daniel-Ribeiro CT. Long-term effect of uncomplicated Plasmodium berghei ANKA malaria on memory and anxiety-like behaviour in C57BL/6 mice. Parasit Vectors 2018; 11:191. [PMID: 29554958 PMCID: PMC5859440 DOI: 10.1186/s13071-018-2778-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 03/06/2018] [Indexed: 01/28/2023] Open
Abstract
Background Cerebral malaria, the main complication of Plasmodium falciparum infection in humans, is associated with persistent neurocognitive sequels both in human disease and the murine experimental model. In recent years, cognitive deficits related to uncomplicated (non-cerebral) malaria have also been reported in chronically exposed residents of endemic areas, but not in some murine experimental models of non-cerebral malaria. This study aimed at evaluating the influence of uncomplicated malaria on different behavioural paradigms associated with memory and anxiety-like parameters in a murine model that has the ability to develop cerebral malaria. Methods Plasmodium berghei ANKA-infected and non-infected C57BL/6 mice were used. Development of cerebral malaria was prevented by chloroquine treatment starting on the fourth day of infection. The control group (non-infected mice) were treated with PBS. The effect of uncomplicated malaria infection on locomotor habituation, short and long-term memory and anxious-like behaviour was evaluated 64 days after parasite clearance in assays including open field, object recognition, Y-maze and light/dark tasks. Results Plasmodium berghei ANKA-infected mice showed significant long-lasting disturbances reflected by a long-term memory-related behaviour on open field and object recognition tasks, accompanied by an anxious-like phenotype availed on open field and light-dark tasks. Conclusions Long-term neurocognitive sequels may follow an uncomplicated malaria episode in an experimental model prone to develop cerebral malaria, even if the infection is treated before the appearance of clinical signs of cerebral impairment.
Collapse
Affiliation(s)
- Luciana Pereira de Sousa
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) da Fundação Oswaldo Cruz (Fiocruz) e da Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro, Brazil.
| | - Roberto Farina de Almeida
- Instituto de Ciências Básicas da Saúde, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Flávia Lima Ribeiro-Gomes
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) da Fundação Oswaldo Cruz (Fiocruz) e da Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro, Brazil
| | - Leonardo José de Moura Carvalho
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) da Fundação Oswaldo Cruz (Fiocruz) e da Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro, Brazil
| | - Tadeu Mello E Souza
- Instituto de Ciências Básicas da Saúde, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Diogo Onofre Gomes de Souza
- Instituto de Ciências Básicas da Saúde, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) da Fundação Oswaldo Cruz (Fiocruz) e da Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Jacobs EG, Weiss B, Makris N, Whitfield-Gabrieli S, Buka SL, Klibanski A, Goldstein JM. Reorganization of Functional Networks in Verbal Working Memory Circuitry in Early Midlife: The Impact of Sex and Menopausal Status. Cereb Cortex 2018; 27:2857-2870. [PMID: 27178194 DOI: 10.1093/cercor/bhw127] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Converging preclinical and human evidence indicates that the decline in ovarian estradiol production during the menopausal transition may play a mechanistic role in the neuronal changes that occur early in the aging process. Here, we present findings from a population-based fMRI study characterizing regional and network-level differences in working memory (WM) circuitry in midlife men and women (N = 142; age range 46-53), as a function of sex and reproductive stage. Reproductive histories and hormonal evaluations were used to determine menopausal status. Participants performed a verbal WM task during fMRI scanning. Results revealed robust differences in task-evoked responses in dorsolateral prefrontal cortex and hippocampus as a function of women's reproductive stage, despite minimal variance in chronological age. Sex differences in regional activity and functional connectivity that were pronounced between men and premenopausal women were diminished for postmenopausal women. Critically, analyzing data without regard to sex or reproductive status obscured group differences in the circuit-level neural strategies associated with successful working memory performance. These findings underscore the importance of reproductive age and hormonal status, over and above chronological age, for understanding sex differences in the aging of memory circuitry. Further, these findings suggest that early changes in working memory circuitry are evident decades before the age range typically targeted in cognitive aging studies.
Collapse
Affiliation(s)
- Emily G Jacobs
- Division of Women's Health, Department of Medicine.,Department of Psychiatry, Brigham and Women's Hospital, Boston, MA 02120, USA.,Harvard Medical School, Boston, MA 02120, USA.,Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Blair Weiss
- Department of Psychiatry, Brigham and Women's Hospital, Boston, MA 02120, USA
| | - Nikos Makris
- Harvard Medical School, Boston, MA 02120, USA.,Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Sue Whitfield-Gabrieli
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stephen L Buka
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| | - Anne Klibanski
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jill M Goldstein
- Division of Women's Health, Department of Medicine.,Department of Psychiatry, Brigham and Women's Hospital, Boston, MA 02120, USA.,Harvard Medical School, Boston, MA 02120, USA.,Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| |
Collapse
|
12
|
Sheppard PAS, Koss WA, Frick KM, Choleris E. Rapid actions of oestrogens and their receptors on memory acquisition and consolidation in females. J Neuroendocrinol 2018; 30. [PMID: 28489296 PMCID: PMC6543823 DOI: 10.1111/jne.12485] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 12/20/2022]
Abstract
Increased attention has been paid in recent years to the ways in which oestrogens and oestrogen receptors rapidly affect learning and memory. These rapid effects occur within a timeframe that is too narrow for the classical genomic mode of action of oestrogen, thus suggesting nonclassical effects as underlying mechanisms. The present review examines recent developments in the study of the rapid effects of 17β-oestradiol and oestrogen receptor (ER) agonists on learning and memory tasks in female rodents, including social recognition, object recognition, object placement (spatial memory) and social learning. By comparing studies utilising systemic or intracranial treatments, as well as pre- and post-acquisition administration of oestradiol or ER agonists, the respective contributions of individual ERs within specific brain regions to various forms of learning and memory can be determined. The first part of this review explores the effects of systemic administration of 17β-oestradiol and ER agonists on memory when administered either pre- or post-acquisition. The second part not only focuses on the effects of pre- and post-acquisition infusions of 17β-oestradiol or ER agonists into the dorsal hippocampus on memory, but also discusses the contributions of other brain regions, including the medial amygdala, medial prefrontal cortex and paraventricular nucleus of the hypothalamus. The cellular mechanisms mediating the rapid effects of 17β-oestradiol on memory, including activation of intracellular signalling cascades and epigenetic processes, are discussed. Finally, the review concludes by comparing pre- and post-acquisition findings and effects of 17β-oestradiol and ER agonists in different brain regions.
Collapse
Affiliation(s)
- P A S Sheppard
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - W A Koss
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - K M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - E Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
13
|
Ikeda MZ, Krentzel AA, Oliver TJ, Scarpa GB, Remage-Healey L. Clustered organization and region-specific identities of estrogen-producing neurons in the forebrain of Zebra Finches (Taeniopygia guttata). J Comp Neurol 2017; 525:3636-3652. [PMID: 28758205 PMCID: PMC6035364 DOI: 10.1002/cne.24292] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/18/2017] [Accepted: 07/20/2017] [Indexed: 01/03/2023]
Abstract
A fast, neuromodulatory role for estrogen signaling has been reported in many regions of the vertebrate brain. Regional differences in the cellular distribution of aromatase (estrogen synthase) in several species suggest that mechanisms for neuroestrogen signaling differ between and even within brain regions. A more comprehensive understanding of neuroestrogen signaling depends on characterizing the cellular identities of neurons that express aromatase. Calcium-binding proteins such as parvalbumin and calbindin are molecular markers for interneuron subtypes, and are co-expressed with aromatase in human temporal cortex. Songbirds like the zebra finch have become important models to understand the brain synthesis of steroids like estrogens and the implications for neurobiology and behavior. Here, we investigated the regional differences in cytoarchitecture and cellular identities of aromatase-expressing neurons in the auditory and sensorimotor forebrain of zebra finches. Aromatase was co-expressed with parvalbumin in the caudomedial nidopallium (NCM) and HVC shelf (proper name) but not in the caudolateral nidopallium (NCL) or hippocampus. By contrast, calbindin was not co-expressed with aromatase in any region investigated. Notably, aromatase-expressing neurons were found in dense somato-somatic clusters, suggesting a coordinated release of local neuroestrogens from clustered neurons. Aromatase clusters were also more abundant and tightly packed in the NCM of males as compared to females. Overall, this study provides new insights into neuroestrogen regulation at the network level, and extends previous findings from human cortex by identifying a subset of aromatase neurons as putative inhibitory interneurons.
Collapse
Affiliation(s)
- Maaya Z Ikeda
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, Massachusetts
| | - Amanda A Krentzel
- Neuroscience and Behavior Program, University of Massachusetts, Amherst, Massachusetts
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, Massachusetts
| | - Tessa J Oliver
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, Massachusetts
| | - Garrett B Scarpa
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, Massachusetts
| | - Luke Remage-Healey
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts
- Neuroscience and Behavior Program, University of Massachusetts, Amherst, Massachusetts
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, Massachusetts
| |
Collapse
|
14
|
Impact of Sex and Menopausal Status on Episodic Memory Circuitry in Early Midlife. J Neurosci 2016; 36:10163-73. [PMID: 27683911 DOI: 10.1523/jneurosci.0951-16.2016] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/08/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Cognitive neuroscience of aging studies traditionally target participants age 65 and older. However, epidemiological surveys show that many women report increased forgetfulness earlier in the aging process, as they transition to menopause. In this population-based fMRI study, we stepped back by over a decade to characterize the changes in memory circuitry that occur in early midlife, as a function of sex and women's reproductive stage. Participants (N = 200; age range, 45-55) performed a verbal encoding task during fMRI scanning. Reproductive histories and serologic evaluations were used to determine menopausal status. Results revealed a pronounced impact of reproductive stage on task-evoked hippocampal responses, despite minimal difference in chronological age. Next, we examined the impact of sex and reproductive stage on functional connectivity across task-related brain regions. Postmenopausal women showed enhanced bilateral hippocampal connectivity relative to premenopausal and perimenopausal women. Across women, lower 17β-estradiol concentrations were related to more pronounced alterations in hippocampal connectivity and poorer performance on a subsequent memory retrieval task, strongly implicating sex steroids in the regulation of this circuitry. Finally, subgroup analyses revealed that high-performing postmenopausal women (relative to low and middle performers) exhibited a pattern of brain activity akin to premenopausal women. Together, these findings underscore the importance of considering reproductive stage, not simply chronological age, to identify neuronal and cognitive changes that unfold in the middle decades of life. In keeping with preclinical studies, these human findings suggest that the decline in ovarian estradiol production during menopause plays a significant role in shaping memory circuitry. SIGNIFICANCE STATEMENT Maintaining intact memory function with age is one of the greatest public health challenges of our time, and women have an increased risk for memory disorders relative to men later in life. We studied adults early in the aging process, as women transition into menopause, to identify neuronal and cognitive changes that unfold in the middle decades of life. Results demonstrate regional and network-level differences in memory encoding-related activity as a function of women's reproductive stage, independent of chronological age. Analyzing data without regard to sex or menopausal status obscured group differences in circuit-level neural strategies associated with successful memory retrieval. These findings suggest that early changes in memory circuitry are evident decades before the age range traditionally targeted by cognitive neuroscience of aging studies.
Collapse
|
15
|
Tuscher JJ, Szinte JS, Starrett JR, Krentzel AA, Fortress AM, Remage-Healey L, Frick KM. Inhibition of local estrogen synthesis in the hippocampus impairs hippocampal memory consolidation in ovariectomized female mice. Horm Behav 2016; 83:60-67. [PMID: 27178577 PMCID: PMC4915975 DOI: 10.1016/j.yhbeh.2016.05.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/02/2016] [Indexed: 01/29/2023]
Abstract
The potent estrogen 17β-Estradiol (E2) plays a critical role in mediating hippocampal function, yet the precise mechanisms through which E2 enhances hippocampal memory remain unclear. In young adult female rodents, the beneficial effects of E2 on memory are generally attributed to ovarian-synthesized E2. However, E2 is also synthesized in the adult brain in numerous species, where it regulates synaptic plasticity and is synthesized in response to experiences such as exposure to females or conspecific song. Although de novo E2 synthesis has been demonstrated in rodent hippocampal cultures, little is known about the functional role of local E2 synthesis in mediating hippocampal memory function. Therefore, the present study examined the role of hippocampal E2 synthesis in hippocampal memory consolidation. Using bilateral dorsal hippocampal infusions of the aromatase inhibitor letrozole, we first found that blockade of dorsal hippocampal E2 synthesis impaired hippocampal memory consolidation. We next found that elevated levels of E2 in the dorsal hippocampus observed 30min after object training were blocked by dorsal hippocampal infusion of letrozole, suggesting that behavioral experience increases acute and local E2 synthesis. Finally, aromatase inhibition did not prevent exogenous E2 from enhancing hippocampal memory consolidation, indicating that hippocampal E2 synthesis is not necessary for exogenous E2 to enhance hippocampal memory. Combined, these data are consistent with the hypothesis that hippocampally-synthesized E2 is necessary for hippocampus-dependent memory consolidation in rodents.
Collapse
Affiliation(s)
- Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Julia S Szinte
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Joseph R Starrett
- Department of Psychological and Brain Sciences and Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, MA, United States
| | - Amanda A Krentzel
- Department of Psychological and Brain Sciences and Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, MA, United States
| | - Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Luke Remage-Healey
- Department of Psychological and Brain Sciences and Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, MA, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
16
|
Luine V. Estradiol: Mediator of memories, spine density and cognitive resilience to stress in female rodents. J Steroid Biochem Mol Biol 2016; 160:189-95. [PMID: 26241030 PMCID: PMC4734902 DOI: 10.1016/j.jsbmb.2015.07.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/28/2015] [Accepted: 07/30/2015] [Indexed: 01/05/2023]
Abstract
Estradiol rapidly activates, within minutes, various physiological functions and behaviors including cognition in rodents. This review describes rapid effects of estradiol on hippocampal dependent learning and memory tasks in rodents. Mechanisms underlying the memory enhancements including the activation of signaling molecules and the enhancement of dendritic spinogenesis are briefly reviewed. In addition, the role of estradiol in the cognitive resilience to chronic stress exhibited only in females is discussed including contributions of ovarian as well as intra-hippocampally derived estrogens to this sex difference. Finally, speculations on possible physiologic functions for rapid mnemonic changes mediated by estrogens are made. Overall, the emergence of a novel and powerful mechanism for regulation of cognition by estradiol is described.
Collapse
Affiliation(s)
- Victoria Luine
- Department of Psychology, Hunter College of CUNY, 695 Park Ave., Rm 611 HN, New York, NY 10065, United States.
| |
Collapse
|
17
|
Frederick AL, Yano H, Trifilieff P, Vishwasrao HD, Biezonski D, Mészáros J, Sibley DR, Kellendonk C, Sonntag KC, Graham DL, Colbran RJ, Stanwood GD, Javitch JA, Javitch JA. Evidence against dopamine D1/D2 receptor heteromers. Mol Psychiatry 2015; 20:1373-85. [PMID: 25560761 PMCID: PMC4492915 DOI: 10.1038/mp.2014.166] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/24/2014] [Accepted: 10/27/2014] [Indexed: 02/07/2023]
Abstract
Hetero-oligomers of G-protein-coupled receptors have become the subject of intense investigation, because their purported potential to manifest signaling and pharmacological properties that differ from the component receptors makes them highly attractive for the development of more selective pharmacological treatments. In particular, dopamine D1 and D2 receptors have been proposed to form hetero-oligomers that couple to Gαq proteins, and SKF83959 has been proposed to act as a biased agonist that selectively engages these receptor complexes to activate Gαq and thus phospholipase C. D1/D2 heteromers have been proposed as relevant to the pathophysiology and treatment of depression and schizophrenia. We used in vitro bioluminescence resonance energy transfer, ex vivo analyses of receptor localization and proximity in brain slices, and behavioral assays in mice to characterize signaling from these putative dimers/oligomers. We were unable to detect Gαq or Gα11 protein coupling to homomers or heteromers of D1 or D2 receptors using a variety of biosensors. SKF83959-induced locomotor and grooming behaviors were eliminated in D1 receptor knockout (KO) mice, verifying a key role for D1-like receptor activation. In contrast, SKF83959-induced motor responses were intact in D2 receptor and Gαq KO mice, as well as in knock-in mice expressing a mutant Ala(286)-CaMKIIα that cannot autophosphorylate to become active. Moreover, we found that, in the shell of the nucleus accumbens, even in neurons in which D1 and D2 receptor promoters are both active, the receptor proteins are segregated and do not form complexes. These data are not compatible with SKF83959 signaling through Gαq or through a D1/D2 heteromer and challenge the existence of such a signaling complex in the adult animals that we used for our studies.
Collapse
Affiliation(s)
- Aliya L. Frederick
- Neuroscience Graduate Program, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Hideaki Yano
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Pierre Trifilieff
- Nutrition and Integrative Neurobiology, INRA UMR 1286; University of Bordeaux, F-33076, Bordeaux, France,Center for Neuroscience. Columbia University, Kolb Research Building, New York, NY10032, USA
| | - Harshad D. Vishwasrao
- Center for Neuroscience. Columbia University, Kolb Research Building, New York, NY10032, USA
| | - Dominik Biezonski
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - József Mészáros
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - David R. Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Christoph Kellendonk
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York, USA,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Kai C. Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Devon L. Graham
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Roger J. Colbran
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA,Vanderbilt Kennedy Center and Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Gregg D. Stanwood
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA,Vanderbilt Kennedy Center and Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jonathan A. Javitch
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, New York, USA,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - J A Javitch
- Departments of Psychiatry and Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
18
|
Blair JA, Bhatta S, McGee H, Casadesus G. Luteinizing hormone: Evidence for direct action in the CNS. Horm Behav 2015; 76:57-62. [PMID: 26172857 PMCID: PMC4741372 DOI: 10.1016/j.yhbeh.2015.06.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 01/09/2023]
Abstract
This article is part of a Special Issue "SBN 2014". Hormonal dysfunction due to aging, especially during menopause, plays a substantial role in cognitive decline as well as the progression and development of neurodegenerative diseases. The hypothalamic-pituitary-gonadal (HPG) axis has long been implicated in changes in behavior and neuronal morphology. Most notably, estrogens have proven beneficial in the healthy brain through a host of different mechanisms. Recently, luteinizing hormone (LH) has emerged as a candidate for further investigation for its role in the CNS. The basis of this is that both LH and the LH receptor are expressed in the brain, and serum levels of LH correlate with cognitive deficits and Alzheimer's disease (AD) incidence. The study of LH in cognition and AD primarily focuses on evaluating the effects of downregulation of this peptide. This literature has shown that decreasing peripheral LH, through a variety of pharmacological interventions, reduces cognitive deficits in ovariectomy and AD models. However, few studies have researched the direct actions of LH on neurons and glial cells. Here we summarize the role of luteinizing hormone in modulating cognition, and we propose a mechanism that underlies a role for brain LH in this process.
Collapse
Affiliation(s)
- Jeffrey A Blair
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Sabina Bhatta
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Henry McGee
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
19
|
Slominski AT, Manna PR, Tuckey RC. On the role of skin in the regulation of local and systemic steroidogenic activities. Steroids 2015; 103:72-88. [PMID: 25988614 PMCID: PMC4631694 DOI: 10.1016/j.steroids.2015.04.006] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/21/2015] [Accepted: 04/21/2015] [Indexed: 01/08/2023]
Abstract
The mammalian skin is a heterogeneous organ/tissue covering our body, showing regional variations and endowed with neuroendocrine activities. The latter is represented by its ability to produce and respond to neurotransmitters, neuropeptides, hormones and neurohormones, of which expression and phenotypic activities can be modified by ultraviolet radiation, chemical and physical factors, as well as by cytokines. The neuroendocrine contribution to the responses of skin to stress is served, in part, by local synthesis of all elements of the hypothalamo-pituitary-adrenal axis. Skin with subcutis can also be classified as a steroidogenic tissue because it expresses the enzyme, CYP11A1, which initiates steroid synthesis by converting cholesterol to pregnenolone, as in other steroidogenic tissues. Pregnenolone, or steroidal precursors from the circulation, are further transformed in the skin to corticosteroids or sex hormones. Furthermore, in the skin CYP11A1 acts on 7-dehydrocholesterol with production of 7-dehydropregnolone, which can be further metabolized to other Δ7steroids, which after exposure to UVB undergo photochemical transformation to vitamin D like compounds with a short side chain. Vitamin D and lumisterol, produced in the skin after exposure to UVB, are also metabolized by CYP11A1 to several hydroxyderivatives. Vitamin D hydroxyderivatives generated by action of CYP11A1 are biologically active and are subject to further hydroxylations by CYP27B1, CYP27A1 and CP24A. Establishment of which intermediates are produced in the epidermis in vivo and whether they circulate on the systemic level represent a future research challenge. In summary, skin is a neuroendocrine organ endowed with steroid/secosteroidogenic activities.
Collapse
Affiliation(s)
- Andrzej T Slominski
- Department of Dermatology, University of Alabama at Birmingham, VA Medical Center, Birmingham, AL, USA.
| | - Pulak R Manna
- Department of immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Robert C Tuckey
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
20
|
Wroolie TE, Kenna HA, Williams KE, Rasgon NL. Cognitive Effects of Hormone Therapy Continuation or Discontinuation in a Sample of Women at Risk for Alzheimer Disease. Am J Geriatr Psychiatry 2015; 23. [PMID: 26209223 PMCID: PMC4654994 DOI: 10.1016/j.jagp.2015.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Use of estrogen-based hormone therapy (HT) as a protection from cognitive decline and Alzheimer disease (AD) is controversial, although cumulative data support HT use when initiated close to menopause onset with estrogen formulations containing 17β-estradiol preferable to conjugated equine estrogen formulations. Little is known regarding specific populations of women who may derive benefit from HT. METHODS Women with heightened risk for AD (aged 49-69), all of whom were taking HT for at least 1 year and most of whom initiated HT close to menopause onset, underwent cognitive assessment followed by randomization to continue or discontinue HT. Assessments were repeated at 2 years after randomization. RESULTS Women who continued HT performed better on cognitive domains composed of measures of verbal memory and combined attention, working memory, and processing speed measures. Women who used 17β-estradiol versus conjugated equine estrogen, whether randomized to continue or discontinue HT, showed better verbal memory performance at the 2-year follow-up assessment. An interaction was also found with HT randomization and family history of AD in a first-degree relative. All female offspring of patients with AD declined in verbal memory; however, women who continued HT declined less than women who discontinued HT. Women without a first-degree relative with AD showed verbal memory improvement (likely because of practice effects) with continuance and declined with discontinuance of HT. CONCLUSION Continuation of HT use appears to protect cognition in women with heightened risk for AD when initiated close to menopause onset.
Collapse
Affiliation(s)
- Tonita E. Wroolie
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, 401 Quarry Road, Stanford University School of Medicine, Stanford, CA 94303-5723, USA
| | - Heather A. Kenna
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, 401 Quarry Road, Stanford University School of Medicine, Stanford, CA 94303-5723, USA
| | - Katherine E. Williams
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, 401 Quarry Road, Stanford University School of Medicine, Stanford, CA 94303-5723, USA
| | - Natalie L. Rasgon
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, 401 Quarry Road, Stanford University School of Medicine, Stanford, CA 94303-5723, USA
| |
Collapse
|
21
|
Frick KM, Kim J, Tuscher JJ, Fortress AM. Sex steroid hormones matter for learning and memory: estrogenic regulation of hippocampal function in male and female rodents. Learn Mem 2015; 22:472-93. [PMID: 26286657 PMCID: PMC4561402 DOI: 10.1101/lm.037267.114] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/09/2015] [Indexed: 01/24/2023]
Abstract
Ample evidence has demonstrated that sex steroid hormones, such as the potent estrogen 17β-estradiol (E2), affect hippocampal morphology, plasticity, and memory in male and female rodents. Yet relatively few investigators who work with male subjects consider the effects of these hormones on learning and memory. This review describes the effects of E2 on hippocampal spinogenesis, neurogenesis, physiology, and memory, with particular attention paid to the effects of E2 in male rodents. The estrogen receptors, cell-signaling pathways, and epigenetic processes necessary for E2 to enhance memory in female rodents are also discussed in detail. Finally, practical considerations for working with female rodents are described for those investigators thinking of adding females to their experimental designs.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Jaekyoon Kim
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| |
Collapse
|
22
|
Frankfurt M, Luine V. The evolving role of dendritic spines and memory: Interaction(s) with estradiol. Horm Behav 2015; 74:28-36. [PMID: 25993604 PMCID: PMC4573337 DOI: 10.1016/j.yhbeh.2015.05.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/21/2015] [Accepted: 05/11/2015] [Indexed: 11/16/2022]
Abstract
This article is part of a Special Issue "Estradiol and Cognition". Memory processing is presumed to depend on synaptic plasticity, which appears to have a role in mediating the acquisition, consolidation, and retention of memory. We have studied the relationship between estrogen, recognition memory, and dendritic spine density in the hippocampus and medial prefrontal cortex, areas critical for memory, across the lifespan in female rodents. The present paper reviews the literature on dendritic spine plasticity in mediating both short and long term memory, as well as the decreased memory that occurs with aging and Alzheimer's disease. It also addresses the role of acute and chronic estrogen treatments in these processes.
Collapse
Affiliation(s)
- Maya Frankfurt
- Department of Science Education, Hofstra-North Shore LIJ School of Medicine, USA.
| | | |
Collapse
|
23
|
Shum C, Macedo SC, Warre-Cornish K, Cocks G, Price J, Srivastava DP. Utilizing induced pluripotent stem cells (iPSCs) to understand the actions of estrogens in human neurons. Horm Behav 2015; 74:228-42. [PMID: 26143621 PMCID: PMC4579404 DOI: 10.1016/j.yhbeh.2015.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 06/11/2015] [Accepted: 06/25/2015] [Indexed: 01/05/2023]
Abstract
This article is part of a Special Issue "Estradiol and Cognition". Over recent years tremendous progress has been made towards understanding the molecular and cellular mechanism by which estrogens exert enhancing effects on cognition, and how they act as a neuroprotective or neurotrophic agent in disease. Currently, much of this work has been carried out in animal models with only a limited number of studies using native human tissue or cells. Recent advances in stem cell technology now make it possible to reprogram somatic cells from humans into induced pluripotent stem cells (iPSCs), which can subsequently be differentiated into neurons of specific lineages. Importantly, the reprogramming of cells allows for the generation of iPSCs that retain the genetic "makeup" of the donor. Therefore, it is possible to generate iPSC-derived neurons from patients diagnosed with specific diseases, that harbor the complex genetic background associated with the disorder. Here, we review the iPSC technology and how it's currently being used to model neural development and neurological diseases. Furthermore, we explore whether this cellular system could be used to understand the role of estrogens in human neurons, and present preliminary data in support of this. We further suggest that the use of iPSC technology offers a novel system to not only further understand estrogens' effects in human cells, but also to investigate the mechanism by which estrogens are beneficial in disease. Developing a greater understanding of these mechanisms in native human cells will also aid in the development of safer and more effective estrogen-based therapeutics.
Collapse
Affiliation(s)
- Carole Shum
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Sara C Macedo
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK; Faculty of Engineering, Universidade do Porto, 4200-465 Porto, Portugal
| | - Katherine Warre-Cornish
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Graham Cocks
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Jack Price
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK.
| |
Collapse
|
24
|
Ervin KSJ, Lymer JM, Matta R, Clipperton-Allen AE, Kavaliers M, Choleris E. Estrogen involvement in social behavior in rodents: Rapid and long-term actions. Horm Behav 2015; 74:53-76. [PMID: 26122289 DOI: 10.1016/j.yhbeh.2015.05.023] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/16/2015] [Accepted: 05/26/2015] [Indexed: 12/21/2022]
Abstract
This article is part of a Special Issue ("Estradiol and cognition"). Estrogens have repeatedly been shown to influence a wide array of social behaviors, which in rodents are predominantly olfactory-mediated. Estrogens are involved in social behavior at multiple levels of processing, from the detection and integration of socially relevant olfactory information to more complex social behaviors, including social preferences, aggression and dominance, and learning and memory for social stimuli (e.g. social recognition and social learning). Three estrogen receptors (ERs), ERα, ERβ, and the G protein-coupled ER 1 (GPER1), differently affect these behaviors. Social recognition, territorial aggression, and sexual preferences and mate choice, all requiring the integration of socially related olfactory information, seem to primarily involve ERα, with ERβ playing a lesser, modulatory role. In contrast, social learning consistently responds differently to estrogen manipulations than other social behaviors. This suggests differential ER involvement in brain regions important for specific social behaviors, such as the ventromedial and medial preoptic nuclei of the hypothalamus in social preferences and aggression, the medial amygdala and hippocampus in social recognition, and the prefrontal cortex and hippocampus in social learning. While the long-term effects of ERα and ERβ on social behavior have been extensively investigated, our knowledge of the rapid, non-genomic, effects of estrogens is more limited and suggests that they may mediate some social behaviors (e.g. social learning) differently from long-term effects. Further research is required to compare ER involvement in regulating social behavior in male and female animals, and to further elucidate the roles of the more recently described G protein-coupled ERs, both the GPER1 and the Gq-mER.
Collapse
Affiliation(s)
- Kelsy S J Ervin
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, Ontario, Canada
| | - Jennifer M Lymer
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, Ontario, Canada
| | - Richard Matta
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, Ontario, Canada
| | | | - Martin Kavaliers
- Department of Psychology, University of Western Ontario, London, Ontario, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
25
|
Frick KM. Molecular mechanisms underlying the memory-enhancing effects of estradiol. Horm Behav 2015; 74:4-18. [PMID: 25960081 PMCID: PMC4573242 DOI: 10.1016/j.yhbeh.2015.05.001] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/25/2015] [Accepted: 05/01/2015] [Indexed: 11/18/2022]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Since the publication of the 1998 special issue of Hormones and Behavior on estrogens and cognition, substantial progress has been made towards understanding the molecular mechanisms through which 17β-estradiol (E2) regulates hippocampal plasticity and memory. Recent research has demonstrated that rapid effects of E2 on hippocampal cell signaling, epigenetic processes, and local protein synthesis are necessary for E2 to facilitate the consolidation of object recognition and spatial memories in ovariectomized female rodents. These effects appear to be mediated by non-classical actions of the intracellular estrogen receptors ERα and ERβ, and possibly by membrane-bound ERs such as the G-protein-coupled estrogen receptor (GPER). New findings also suggest a key role of hippocampally-synthesized E2 in regulating hippocampal memory formation. The present review discusses these findings in detail and suggests avenues for future study.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 E. Hartford Ave., Milwaukee, WI 53211, USA.
| |
Collapse
|
26
|
Brandt N, Vierk R, Fester L, Zhou L, Imholz P, Rune GM. [Gender and the effects of steroid hormones in the central nervous system]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2015; 57:1054-60. [PMID: 25091372 DOI: 10.1007/s00103-014-2014-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Degenerative diseases of the central nervous system, the incidence and prevalence of which vary between men and women, often manifest in the hippocampus. Neurosteroids are hormones that are synthesized in the CNS, and it is here that they exert their influence. Estrogen and testosterone are examples of neurosteroid hormones. In the hippocampus, an area of the brain closely associated with learning and memory, the local synthesis of estrogen in females, but not in males, is essential for the plasticity and stability of the synapses. The inhibition of estrogen synthesis in the female hippocampus causes a reduction in long-term potentiation (LTP), an electrophysiological parameter of learning and memory, thus resulting in a significant loss of synapses. In light of this, the fact that estrogen has been attributed with many neuroprotective functions in degenerative diseases of the CNS suggests that therapeutic concepts involving the use of estrogen are possibly only effective in women, but not in men. These findings similarly provide a basis for explaining the gender dimorphism that has been found in certain degenerative illnesses of the CNS.
Collapse
Affiliation(s)
- N Brandt
- Institut für Neuroanatomie, Universitätsklinikum Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Deutschland
| | | | | | | | | | | |
Collapse
|
27
|
Zhao XS, Fu WY, Hung KW, Chien WWY, Li Z, Fu AK, Ip NY. NRC-interacting factor directs neurite outgrowth in an activity-dependent manner. Neuroscience 2015; 289:207-13. [PMID: 25573434 DOI: 10.1016/j.neuroscience.2014.12.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/05/2014] [Accepted: 12/24/2014] [Indexed: 11/18/2022]
Abstract
Nuclear hormone receptor coregulator-interacting factor 1 (NIF-1) is a zinc finger nuclear protein that was initially identified to enhance nuclear hormone receptor transcription via its interaction with nuclear hormone receptor coregulator (NRC). NIF-1 may regulate gene transcription either by modulating general transcriptional machinery or remodeling chromatin structure through interactions with specific protein partners. We previously reported that the cytoplasmic/nuclear localization of NIF-1 is regulated by the neuronal Cdk5 activator p35, suggesting potential neuronal functions for NIF-1. The present study reveals that NIF-1 plays critical roles in regulating neuronal morphogenesis at early stages. NIF-1 was prominently expressed in the nuclei of developing rat cortical neurons. Knockdown of NIF-1 expression attenuated both neurite outgrowth in cultured cortical neurons and retinoic acid (RA)-treated Neuro-2a neuroblastoma cells. Furthermore, activity-induced Ca(2+) influx, which is critical for neuronal morphogenesis, stimulated the nuclear localization of NIF-1 in cortical neurons. Suppression of NIF-1 expression reduced the up-regulation of neuronal activity-dependent gene transcription. These findings collectively suggest that NIF-1 directs neuronal morphogenesis during early developmental stages through modulating activity-dependent gene transcription.
Collapse
Affiliation(s)
- X-S Zhao
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - W-Y Fu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - K-W Hung
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - W W Y Chien
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Z Li
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - A K Fu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - N Y Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.
| |
Collapse
|
28
|
Fortress AM, Frick KM. Epigenetic regulation of estrogen-dependent memory. Front Neuroendocrinol 2014; 35:530-49. [PMID: 24878494 PMCID: PMC4174980 DOI: 10.1016/j.yfrne.2014.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 02/09/2023]
Abstract
Hippocampal memory formation is highly regulated by post-translational histone modifications and DNA methylation. Accordingly, these epigenetic processes play a major role in the effects of modulatory factors, such as sex steroid hormones, on hippocampal memory. Our laboratory recently demonstrated that the ability of the potent estrogen 17β-estradiol (E2) to enhance hippocampal-dependent novel object recognition memory in ovariectomized female mice requires ERK-dependent histone H3 acetylation and DNA methylation in the dorsal hippocampus. Although these data provide valuable insight into the chromatin modifications that mediate the memory-enhancing effects of E2, epigenetic regulation of gene expression is enormously complex. Therefore, more research is needed to fully understand how E2 and other hormones employ epigenetic alterations to shape behavior. This review discusses the epigenetic alterations shown thus far to regulate hippocampal memory, briefly reviews the effects of E2 on hippocampal function, and describes in detail our work on epigenetic regulation of estrogenic memory enhancement.
Collapse
Affiliation(s)
- Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
29
|
Yoder KM, Phan ML, Lu K, Vicario DS. He hears, she hears: are there sex differences in auditory processing? Dev Neurobiol 2014; 75:302-14. [PMID: 25220950 DOI: 10.1002/dneu.22231] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/20/2014] [Accepted: 09/11/2014] [Indexed: 01/18/2023]
Abstract
Songbirds learn individually unique songs through vocal imitation and use them in courtship and territorial displays. Previous work has identified a forebrain auditory area, the caudomedial nidopallium (NCM), that appears specialized for discriminating and remembering conspecific vocalizations. In zebra finches (ZFs), only males produce learned vocalizations, but both sexes process these and other signals. This study assessed sex differences in auditory processing by recording extracellular multiunit activity at multiple sites within NCM. Juvenile female ZFs (n = 46) were reared in individual isolation and artificially tutored with song. In adulthood, songs were played back to assess auditory responses, stimulus-specific adaptation, neural bias for conspecific song, and memory for the tutor's song, as well as recently heard songs. In a subset of females (n = 36), estradiol (E2) levels were manipulated to test the contribution of E2, known to be synthesized in the brain, to auditory responses. Untreated females (n = 10) showed significant differences in response magnitude and stimulus-specific adaptation compared to males reared in the same paradigm (n = 9). In hormone-manipulated females, E2 augmentation facilitated the memory for recently heard songs in adulthood, but neither E2 augmentation (n = 15) nor E2 synthesis blockade (n = 9) affected tutor song memory or the neural bias for conspecific song. The results demonstrate subtle sex differences in processing communication signals, and show that E2 levels in female songbirds can affect the memory for songs of potential suitors, thus contributing to the process of mate selection. The results also have potential relevance to clinical interventions that manipulate E2 in human patients.
Collapse
Affiliation(s)
- Kathleen M Yoder
- Department of Psychological & Brain Sciences, Johns Hopkins University, Baltimore, Maryland, 21218
| | | | | | | |
Collapse
|
30
|
Chao A, Paon A, Remage-Healey L. Dynamic variation in forebrain estradiol levels during song learning. Dev Neurobiol 2014; 75:271-86. [PMID: 25205304 DOI: 10.1002/dneu.22228] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 08/18/2014] [Accepted: 09/02/2014] [Indexed: 12/24/2022]
Abstract
Estrogens shape brain circuits during development, and the capacity to synthesize estrogens locally has consequences for both sexual differentiation and the acute modulation of circuits during early learning. A recently optimized method to detect and quantify fluctuations in brain estrogens in vivo provides a direct means to explore how brain estrogen production contributes to both differentiation and neuromodulation during development. Here, we use this method to test the hypothesis that neuroestrogens are sexually differentiated as well as dynamically responsive to song tutoring (via passive video/audio playback) during the period of song learning in juvenile zebra finches. Our results show that baseline neuroestradiol levels in the caudal forebrain do not differ between males and females during an early critical masculinization window. Instead, we observe a prominent difference between males and females in baseline neuroestradiol that emerges during the subadult stage as animals approach sexual maturity. Second, we observe that fluctuating neuroestradiol levels during periods of passive song tutoring exhibit a markedly different profile in juveniles as compared to adults. Specifically, neuroestrogens in the caudal forebrain are elevated following (rather than during) tutor song exposure in both juvenile males and females, suggesting an important role for the early consolidation of tutor song memories. These results further reveal a circadian influence on the fluctuations in local neuroestrogens during sensory/cognitive tasks. Taken together, these findings uncover several unexpected features of brain estrogen synthesis in juvenile animals that may have implications for secondary masculinization as well as the consolidation of recent sensory experiences.
Collapse
Affiliation(s)
- Andrew Chao
- Neuroscience and Behavior Program, Center for Neuroendocrine Studies, Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, Massachusetts, 01003
| | | | | |
Collapse
|
31
|
Luine VN. Estradiol and cognitive function: past, present and future. Horm Behav 2014; 66:602-18. [PMID: 25205317 PMCID: PMC4318702 DOI: 10.1016/j.yhbeh.2014.08.011] [Citation(s) in RCA: 306] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/26/2014] [Accepted: 08/29/2014] [Indexed: 12/13/2022]
Abstract
A historical perspective on estradiol's enhancement of cognitive function is presented, and research, primarily in animals, but also in humans, is reviewed. Data regarding the mechanisms underlying the enhancements are discussed. Newer studies showing rapid effects of estradiol on consolidation of memory through membrane interactions and activation of inter-cellular signaling pathways are reviewed as well as studies focused on traditional genomic mechanisms. Recent demonstrations of intra-neuronal estradiol synthesis and possible actions as a neurosteroid to promote memory are discussed. This information is applied to the critical issue of the current lack of effective hormonal (or other) treatments for cognitive decline associated with menopause and aging. Finally, the critical period hypothesis for estradiol effects is discussed along with novel strategies for hormone/drug development. Overall, the historical record documents that estradiol positively impacts some aspects of cognitive function, but effective therapeutic interventions using this hormone have yet to be realized.
Collapse
Affiliation(s)
- Victoria N Luine
- Department of Psychology, Hunter College of CUNY, New York, NY, USA.
| |
Collapse
|
32
|
Regulation of object recognition and object placement by ovarian sex steroid hormones. Behav Brain Res 2014; 285:140-57. [PMID: 25131507 DOI: 10.1016/j.bbr.2014.08.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 07/29/2014] [Accepted: 08/01/2014] [Indexed: 12/26/2022]
Abstract
The ovarian hormones 17β-estradiol (E2) and progesterone (P4) are potent modulators of hippocampal memory formation. Both hormones have been demonstrated to enhance hippocampal memory by regulating the cellular and molecular mechanisms thought to underlie memory formation. Behavioral neuroendocrinologists have increasingly used the object recognition and object placement (object location) tasks to investigate the role of E2 and P4 in regulating hippocampal memory formation in rodents. These one-trial learning tasks are ideal for studying acute effects of hormone treatments on different phases of memory because they can be administered during acquisition (pre-training), consolidation (post-training), or retrieval (pre-testing). This review synthesizes the rodent literature testing the effects of E2 and P4 on object recognition (OR) and object placement (OP), and the molecular mechanisms in the hippocampus supporting memory formation in these tasks. Some general trends emerge from the data. Among gonadally intact females, object memory tends to be best when E2 and P4 levels are elevated during the estrous cycle, pregnancy, and in middle age. In ovariectomized females, E2 given before or immediately after testing generally enhances OR and OP in young and middle-aged rats and mice, although effects are mixed in aged rodents. Effects of E2 treatment on OR and OP memory consolidation can be mediated by both classical estrogen receptors (ERα and ERβ), and depend on glutamate receptors (NMDA, mGluR1) and activation of numerous cell signaling cascades (e.g., ERK, PI3K/Akt, mTOR) and epigenetic processes (e.g., histone acetylation, DNA methylation). Acute P4 treatment given immediately after training also enhances OR and OP in young and middle-aged ovariectomized females by activating similar cell signaling pathways as E2 (e.g., ERK, mTOR). The few studies that have administered both hormones in combination suggest that treatment can enhance OR and OP, but that effects are highly dependent on factors such as dose and timing of administration. In addition to providing more detail on these general conclusions, this review will discuss directions for future avenues of research into the hormonal regulation of object memory.
Collapse
|
33
|
Hippocampal estradiol synthesis and its significance for hippocampal synaptic stability in male and female animals. Neuroscience 2014; 274:24-32. [DOI: 10.1016/j.neuroscience.2014.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 04/23/2014] [Accepted: 05/04/2014] [Indexed: 01/18/2023]
|
34
|
Luine V. Recognition memory tasks in neuroendocrine research. Behav Brain Res 2014; 285:158-64. [PMID: 24837746 DOI: 10.1016/j.bbr.2014.04.032] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 04/02/2014] [Accepted: 04/17/2014] [Indexed: 12/26/2022]
Abstract
The recognition memory tasks, novel object and novel object location, have been beneficial to neuroendocrine research concerning the effects of gonadal and adrenal hormones on cognitive function. This review discusses the advantages of these tasks in comparison with other learning and memory tasks. Experiments conducted across a number of laboratories show that gonadal hormones, both estradiol and testosterone, promote memory while the adrenal hormone, corticosterone, impairs memory. The effects of these steroid hormones on spine density in the prefrontal cortex and hippocampus are also briefly presented. Overall, results show that these steroid hormones are potent modulators of memory consolidation in rodent models.
Collapse
Affiliation(s)
- Victoria Luine
- Department of Psychology, Hunter College of CUNY, New York, NY, United States.
| |
Collapse
|
35
|
Lacreuse A, Chang J, Metevier CM, LaClair M, Meyer JS, Ferris CM. Oestradiol modulation of cognition in adult female marmosets (Callithrix jacchus). J Neuroendocrinol 2014; 26:296-309. [PMID: 24617856 PMCID: PMC4040528 DOI: 10.1111/jne.12147] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 02/24/2014] [Accepted: 03/06/2014] [Indexed: 01/03/2023]
Abstract
The common marmoset (Callithrix jacchus) provides many advantages over traditional rodent and macaque species as a model for human ageing and may be very useful for studying the effects of sex steroids on cognitive and brain ageing. We present the first study examining the effects of oestrogens on cognitive function in female marmosets. Adult monkeys (3-5 years of age) were trained to a specific learning criterion on a battery of cognitive tasks preoperatively (object discrimination, delayed response with increasing delays and detour reaching with opaque box) and were tested on different versions of these tasks (object reversals, delayed response with randomised delays and detour reaching with clear box) after ovariectomy and simultaneous implantation with 17β-oestradiol (E2 ) (n = 6) or blank (n = 6) Silastic capsules. Acquisition of a delayed matching-to-position task with a 1-s delay was also administered after completion of these tests. E2 -treated monkeys were significantly impaired on the second reversal and showed an increase in perseverative responding from reversals 1-3. Their performance also tended to be worse than that of control monkeys on the delayed response task. Performance acquisition on the delayed matching-to-position tended to be better in E2 -treated relative to control monkeys, although the group difference did not reach statistical significance. No effect of treatment was detected for detour reaching or affiliative behaviours. Overall, the findings indicate that E2 compromises performance on prefrontally-mediated tasks. The suggestion that E2 may improve acquisition on tasks dependent on the hippocampus will require further validation. These results are discussed in the context of dopaminergic and serotonergic signalling. We conclude that the marmoset is a useful new primate model for examining the effects of oestrogens on cognitive function.
Collapse
Affiliation(s)
- Agnès Lacreuse
- Department of Psychology, University of Massachusetts, Amherst MA 01003
- Correspondence to: Department of Psychology 135 Hicks Way University of Massachusetts Amherst, MA 01003 Phone: 413-545-2183 Fax: 413-545-0996
| | - Jeemin Chang
- Department of Psychology, University of Massachusetts, Amherst MA 01003
| | | | - Matthew LaClair
- Neuroscience and Behavior graduate program, University of Massachusetts, Amherst MA
| | - Jerrold S. Meyer
- Department of Psychology, University of Massachusetts, Amherst MA 01003
| | - Craig M. Ferris
- Department of Psychology, Northeastern University, Boston MA 02115
| |
Collapse
|
36
|
Rasgon NL, Geist CL, Kenna HA, Wroolie TE, Williams KE, Silverman DHS. Prospective randomized trial to assess effects of continuing hormone therapy on cerebral function in postmenopausal women at risk for dementia. PLoS One 2014; 9:e89095. [PMID: 24622517 PMCID: PMC3951184 DOI: 10.1371/journal.pone.0089095] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/13/2014] [Indexed: 01/02/2023] Open
Abstract
The objective of this study was to examine the effects of estrogen-based hormone therapy (HT) on regional cerebral metabolism in postmenopausal women (mean age = 58, SD = 5) at risk for development of dementia. The prospective clinical trial design included pre- and post-intervention neuroimaging of women randomized to continue (HT+) or discontinue (HT−) therapy following an average of 10 years of use. The primary outcome measure was change in brain metabolism during the subsequent two years, as assessed with fluorodeoxyglucose-18 positron emission tomography (FDG-PET). Longitudinal FDG-PET data were available for 45 study completers. Results showed that women randomized to continue HT experienced relative preservation of frontal and parietal cortical metabolism, compared with women randomized to discontinue HT. Women who discontinued 17-β estradiol (17βE)-based HT, as well as women who continued conjugated equine estrogen (CEE)-based HT, exhibited significant decline in metabolism of the precuneus/posterior cingulate cortical (PCC) area. Significant decline in PCC metabolism was additionally seen in women taking concurrent progestins (with either 17βE or CEE). Together, these findings suggest that among postmenopausal subjects at risk for developing dementia, regional cerebral cortical metabolism is relatively preserved for at least two years in women randomized to continue HT, compared with women randomized to discontinue HT. In addition, continuing unopposed 17βE therapy is associated specifically with preservation of metabolism in PCC, known to undergo the most significant decline in the earliest stages of Alzheimer's disease. Trial Registration ClinicalTrials.govNCT00097058
Collapse
Affiliation(s)
- Natalie L. Rasgon
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| | - Cheri L. Geist
- UCLA David Geffen School of Medicine, Department of Molecular and Medical Pharmacology, Ahmanson Translational Imaging Division, University of California Los Angeles School of Medicine, Los Angeles, California, United States of America
| | - Heather A. Kenna
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Tonita E. Wroolie
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Katherine E. Williams
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Daniel H. S. Silverman
- UCLA David Geffen School of Medicine, Department of Molecular and Medical Pharmacology, Ahmanson Translational Imaging Division, University of California Los Angeles School of Medicine, Los Angeles, California, United States of America
| |
Collapse
|
37
|
Frye CA, Walf AA, Kohtz AS, Zhu Y. Progesterone-facilitated lordosis of estradiol-primed mice is attenuated by knocking down expression of membrane progestin receptors in the midbrain. Steroids 2014; 81:17-25. [PMID: 24269738 PMCID: PMC4540077 DOI: 10.1016/j.steroids.2013.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Evidence is emerging of the role of membrane progestin receptors (referred to as mPRs herein: members of Progestin and AdipoQ Receptor (Paqr) family) as a novel brain target in mammals, such as rats. In the present study, the role of mPRs in mice was assessed to further elucidate the conservation of this mechanism across species. The brain target investigated was the midbrain ventral tegmental area (VTA) given its described role for rapid actions of progestins for reproduction. Studies tested the hypothesis that if mPRs are required for progestin-facilitated lordosis through actions in the VTA, then knockdown of mPRs in the VTA will attenuate lordosis. Ovariectomized (OVX) mice were subcutaneously injected with estradiol (E2) and progesterone (P4), and infused with antisense oligodeoxynucleotides (AS-ODNs) to mPRα (Paqr7) and/or mPRβ (Paqr8) or vehicle to the lateral ventricle or VTA. Mice were assessed for reproductive behavior (lordosis and aggression/rejection quotients) in a standard mating task. Results supported our hypothesis. E2+P4-facilitated lordosis was significantly reduced, and aggression/rejection increased, with infusions of mPRα, mPRβ, or mPRαβ AS-ODNs to the lateral ventricle, compared to vehicle. E2+P4-facilitated lordosis was significantly decreased, and aggression/rejection increased, with mPRβ or mPRαβ AS-ODNs to the VTA of C57/BL6 mice. Both mPRɑ and mPRβ AS-ODNs reduced lordosis, and increased aggression/rejection, of wildtype (C57/BL6x129) mice, but not nuclear PR knockout mice. Thus, mPRs may be a novel target of progestins for reproductive behavior of mice.
Collapse
Affiliation(s)
- Cheryl A Frye
- Dept. of Psychology, The University at Albany-SUNY, Albany, NY 12222, USA; Dept. of Biological Sciences, The University at Albany-SUNY, Albany, NY 12222, USA; The Center for Neuroscience, The University at Albany-SUNY, Albany, NY 12222, USA; The Center for Life Science Research, The University at Albany-SUNY, Albany, NY 12222, USA; Dept. of Chemistry, The University of Alaska-Fairbanks, IDeA Network of Biomedical Excellence (INBRE), Fairbanks, AK 99775, USA.
| | - Alicia A Walf
- Dept. of Psychology, The University at Albany-SUNY, Albany, NY 12222, USA; Dept. of Chemistry, The University of Alaska-Fairbanks, IDeA Network of Biomedical Excellence (INBRE), Fairbanks, AK 99775, USA; Cognitive Science Department, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA
| | - Amy S Kohtz
- Dept. of Psychology, The University at Albany-SUNY, Albany, NY 12222, USA
| | - Yong Zhu
- Dept. of Biology, East Carolina University, Greenville, NC 27858-4353, USA
| |
Collapse
|
38
|
Abstract
AbstractEnvironmental enrichment aims to improve the well-being of laboratory animals and provides an opportunity to improve experimental reliability and validity. Animals raised in more stimulating environments have improved learning and memory as well as more complex brain architecture. However, the effects of environmental enrichment on motor performance, anxiety and emotional development have been poorly studied. Moreover, most investigators studying the effects of enrichment provide extremely large and complex housing conditions to maximize the likelihood of finding effects. These situations are difficult to replicate across animal facilities and are not operationally practical. In this experiment, we investigated how simple, inexpensive disposable shelterstyle enrichment items alter behavior in C57Bl/6 and 129S6 mice. Breeding pairs were established in the presence of a Ketchum “Refuge”, Shepherd Shack “Dome”, or no enrichment. Offspring were assessed neurobehaviorally, either just after weaning (pre-adolescent, P22–P25), or as young adults (P60–P90). Major strain differences were observed in open field activity, elevated maze exploration, and Y-maze activity levels. The presence of the Refuge and/or Dome enrichment shelters significantly altered motor activity, coordination and some measures of anxiety. Mice housed in the presence of shelters were also less dominant than control mice in a tube test assay. Our experiments provide a detailed analysis of the effects of inexpensive and practical methods of housing enrichment on biobehavioral phenotypes in these two commonly used strains of laboratory mice, and suggest that the effects of these shelters on mouse neurobiology and behavior need to be rigorously analyzed before being adopted within vivariums.
Collapse
|
39
|
Remage-Healey L, Jeon SD, Joshi NR. Recent evidence for rapid synthesis and action of oestrogens during auditory processing in a songbird. J Neuroendocrinol 2013; 25:1024-31. [PMID: 23746380 PMCID: PMC4153829 DOI: 10.1111/jne.12055] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/13/2013] [Accepted: 06/01/2013] [Indexed: 11/28/2022]
Abstract
It is now clear that oestrogens are not only circulating reproductive hormones, but that they also have neurotransmitter-like properties in a wide range of brain circuits. The view of oestrogens as intrinsic neuromodulators that shape behaviour has been bolstered by a series of recent developments from multiple vertebrate model systems. Here, we review several recent findings from studies of songbirds showing how the identified neural circuits that govern auditory processing and sensorimotor integration are modulated by the local and acute production of oestrogens. First, studies using in vivo microdialysis demonstrate that oestrogens fluctuate in the auditory cortex (30-min time bin resolution) when songbirds are hearing song and interacting with conspecifics. Second, oestrogens rapidly boost the auditory-evoked activity of neurones in the same auditory cortical region, enhancing auditory processing. Third, local pharmacological blockade of oestrogen signalling in this region impairs auditory neuronal responsiveness, as well as behavioural song preferences. Fourth, the rapid actions of oestrogens that occur within the auditory cortex can propagate downstream (trans-synaptically) to sensorimotor circuits to enhance the neural representation of song. Lastly, we present new evidence showing that the receptor for the rapid actions of oestradiol is likely in neuronal membranes, and that traditional nuclear oestrogen receptor agonists do not mimic these rapid actions. Broadly speaking, many of these findings are observed in both males and females, emphasising the fundamental importance of oestrogens in neural circuit function. Together, these and other emergent studies provide support for rapid, brain-derived oestrogen signalling in regulating sensorimotor integration, learning and perception.
Collapse
|
40
|
Ervin KSJ, Phan A, Gabor CS, Choleris E. Rapid oestrogenic regulation of social and nonsocial learning. J Neuroendocrinol 2013; 25:1116-32. [PMID: 23876061 DOI: 10.1111/jne.12079] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 06/26/2013] [Accepted: 07/16/2013] [Indexed: 12/16/2022]
Abstract
Much research on oestrogens has focused on their long-term action, exerting behavioural effects within hours to days through gene transcription. Oestrogens also affect behaviour on a much shorter time scale. These rapid effects are assumed to occur through cell signalling and can elicit a behavioural effect as early as 15 min after treatment. These effects on behaviour have primarily been explored through the action of oestradiol at three well-known oestrogen receptors (ERs): ERα, ERβ and the more recently described G protein-coupled ER1 (GPER1). The rapid effects of oestradiol and ER agonists have been tested on both social and nonsocial learning paradigms. Social learning refers to a paradigm in which an animal acquires information and modifies its behaviour based on observation of another animal, commonly studied using the social transmission of food preferences paradigm. When administered shortly before testing, oestradiol rapidly improves social learning on this task, although no ER agonist has definitive, comparable improving effects. Some evidence points to GPER1, whereas ERα impairs, and ERβ activation has no effect on social learning. Conversely, ERα and GPER1 play a larger role than ERβ in the rapid improving effect of oestrogens on nonsocial learning, including social and object recognition. In addition, when administered immediately post-acquisition, oestrogens also rapidly improve memory consolidation in a variety of learning paradigms: object recognition, object placement, inhibitory avoidance and the Morris water maze, indicating that oestradiol affects the consolidation of multiple types of memory. Evidence suggests that these improvements are the result of oestrogens acting in the dorsal hippocampus where selective activation of all three ERs shows rapid improving effects on spatial learning comparable to oestradiol. However, the hippocampus is not necessary for rapid oestradiol improvements on social recognition. Although acute treatment with oestradiol enhances learning and memory on various social and nonsocial learning paradigms, the specific ERs play different roles in each type of learning. Future research should aim to further determine the roles of ERs with respect to the enhancing effects of oestradiol on learning and memory, and also determine where in the brain oestradiol acts to affect social and nonsocial learning.
Collapse
Affiliation(s)
- K S J Ervin
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, Canada
| | | | | | | |
Collapse
|
41
|
Frick KM. Epigenetics, oestradiol and hippocampal memory consolidation. J Neuroendocrinol 2013; 25:1151-62. [PMID: 24028406 PMCID: PMC3943552 DOI: 10.1111/jne.12106] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 08/13/2013] [Accepted: 09/05/2013] [Indexed: 02/02/2023]
Abstract
Epigenetic alterations of histone proteins and DNA are essential for hippocampal synaptic plasticity and cognitive function, and contribute to the aetiology of psychiatric disorders and neurodegenerative diseases. Hippocampal memory formation depends on histone alterations and DNA methylation, and increasing evidence suggests that the regulation of these epigenetic processes by modulatory factors, such as environmental enrichment, stress and hormones, substantially influences memory function. Recent work from our laboratory suggests that the ability of the sex-steroid hormone 17β-oestradiol (E2 ) to enhance novel object recognition memory consolidation in young adult female mice is dependent on histone H3 acetylation and DNA methylation in the dorsal hippocampus. Our data also suggest that enzymes mediating DNA methylation and histone acetylation work in concert to regulate the effects of E2 on memory consolidation. These findings shed light on the epigenetic mechanisms that influence hormonal modulation of cognitive function, and may have important implications for understanding how hormones influence cognition in adulthood and ageing. The present review provides a brief overview of the literature on epigenetics and memory, describes in detail our findings demonstrating that epigenetic alterations regulate E2 -induced memory enhancement in female mice, and discusses future directions for research on the epigenetic regulation of E2 -induced memory enhancement.
Collapse
Affiliation(s)
- Karyn M. Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211
| |
Collapse
|
42
|
Galea LAM, Wainwright SR, Roes MM, Duarte-Guterman P, Chow C, Hamson DK. Sex, hormones and neurogenesis in the hippocampus: hormonal modulation of neurogenesis and potential functional implications. J Neuroendocrinol 2013; 25:1039-61. [PMID: 23822747 DOI: 10.1111/jne.12070] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/23/2013] [Accepted: 06/29/2013] [Indexed: 12/12/2022]
Abstract
The hippocampus is an area of the brain that undergoes dramatic plasticity in response to experience and hormone exposure. The hippocampus retains the ability to produce new neurones in most mammalian species and is a structure that is targeted in a number of neurodegenerative and neuropsychiatric diseases, many of which are influenced by both sex and sex hormone exposure. Intriguingly, gonadal and adrenal hormones affect the structure and function of the hippocampus differently in males and females. Adult neurogenesis in the hippocampus is regulated by both gonadal and adrenal hormones in a sex- and experience-dependent way. Sex differences in the effects of steroid hormones to modulate hippocampal plasticity should not be completely unexpected because the physiology of males and females is different, with the most notable difference being that females gestate and nurse the offspring. Furthermore, reproductive experience (i.e. pregnancy and mothering) results in permanent changes to the maternal brain, including the hippocampus. This review outlines the ability of gonadal and stress hormones to modulate multiple aspects of neurogenesis (cell proliferation and cell survival) in both male and female rodents. The function of adult neurogenesis in the hippocampus is linked to spatial memory and depression, and the present review provides early evidence of the functional links between the hormonal modulation of neurogenesis that may contribute to the regulation of cognition and stress.
Collapse
Affiliation(s)
- L A M Galea
- Department of Psychology, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Kerr TM, Muller CL, Miah M, Jetter CS, Pfeiffer R, Shah C, Baganz N, Anderson GM, Crawley JN, Sutcliffe JS, Blakely RD, Veenstra-Vanderweele J. Genetic background modulates phenotypes of serotonin transporter Ala56 knock-in mice. Mol Autism 2013; 4:35. [PMID: 24083388 PMCID: PMC3851031 DOI: 10.1186/2040-2392-4-35] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 08/27/2013] [Indexed: 12/25/2022] Open
Abstract
Background Previously, we identified multiple, rare serotonin (5-HT) transporter (SERT) variants in children with autism spectrum disorder (ASD). Although in our study the SERT Ala56 variant was over-transmitted to ASD probands, it was also seen in some unaffected individuals, suggesting that associated ASD risk is influenced by the epistatic effects of other genetic variation. Subsequently, we established that mice expressing the SERT Ala56 variant on a 129S6/S4 genetic background display multiple biochemical, physiological and behavioral changes, including hyperserotonemia, altered 5-HT receptor sensitivity, and altered social, communication, and repetitive behavior. Here we explore the effects of genetic background on SERT Ala56 knock-in phenotypes. Methods To explore the effects of genetic background, we backcrossed SERT Ala56 mice on the 129 background into a C57BL/6 (B6) background to achieve congenic B6 SERT Ala56 mice, and assessed autism-relevant behavior, including sociability, ultrasonic vocalizations, and repetitive behavior in the home cage, as well as serotonergic phenotypes, including whole blood serotonin levels and serotonin receptor sensitivity. Results One consistent phenotype between the two strains was performance in the tube test for dominance, where mutant mice displayed a greater tendency to withdraw from a social encounter in a narrow tube as compared to wildtype littermate controls. On the B6 background, mutant pup ultrasonic vocalizations were significantly increased, in contrast to decreased vocalizations seen previously on the 129 background. Several phenotypes seen on the 129 background were reduced or absent when the mutation was placed on the B6 background, including hyperserotonemia, 5-HT receptor hypersensivity, and repetitive behavior. Conclusions Our findings provide a cogent example of how epistatic interactions can modulate the impact of functional genetic variation and suggest that some aspects of social behavior may be especially sensitive to changes in SERT function. Finally, these results provide a platform for the identification of genes that may modulate the risk of ASD in humans.
Collapse
Affiliation(s)
- Travis M Kerr
- Department of Psychiatry, Vanderbilt University, 465 21st Ave S, Nashville, TN 37232, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Boulware MI, Heisler JD, Frick KM. The memory-enhancing effects of hippocampal estrogen receptor activation involve metabotropic glutamate receptor signaling. J Neurosci 2013; 33:15184-94. [PMID: 24048848 PMCID: PMC6618419 DOI: 10.1523/jneurosci.1716-13.2013] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/22/2013] [Accepted: 08/10/2013] [Indexed: 01/22/2023] Open
Abstract
Our laboratory has demonstrated that 17β-estradiol (E2) enhances hippocampal memory consolidation via rapid activation of multiple intracellular signaling cascades, including the ERK/MAPK cascade (Fernandez et al., 2008; Fan et al., 2010). However, the receptor mechanisms responsible for these effects of E2 remain unclear. In vitro, estrogen receptor α (ERα) signaling through metabotropic glutamate receptor 1a (mGluR1a) leads to ERK-dependent CREB phosphorylation (Boulware et al., 2005), suggesting that interactions between ERs and mGluR1a may be vital to the memory-enhancing effects of E2. As such, the present study tested the roles of classical estrogen receptors (ERα and ERβ) and mGluR1a in mediating the effects of E2 on hippocampal memory consolidation. Dorsal hippocampal (DH) infusion of ERα (PPT) or ERβ (DPN) agonists enhanced novel object recognition and object placement memory in ovariectomized female mice in an ERK-dependent manner, suggesting that these receptors influence memory by rapidly activating hippocampal cell signaling. Next, DH infusion of the mGluR1a antagonist LY367385 blocked the object and spatial memory facilitation induced by E2, PPT, and DPN, demonstrating that ER/mGluR1a signaling is critical for the memory-enhancing effects of E2. Finally, we show that ERα, ERβ, mGluR1, and ERK all reside within specialized membrane microdomains of the DH, and that ERα and ERβ physically interact with mGluR1, providing a means through which ERs may activate mGluRs and downstream signaling. Together, these findings provide the first in vivo evidence demonstrating that ER/mGluR signaling can mediate the beneficial effects of E2 on hippocampal memory consolidation.
Collapse
Affiliation(s)
- Marissa I. Boulware
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211
| | - John D. Heisler
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211
| | - Karyn M. Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211
| |
Collapse
|
45
|
Mennenga S, Bimonte-Nelson H. Translational cognitive endocrinology: designing rodent experiments with the goal to ultimately enhance cognitive health in women. Brain Res 2013; 1514:50-62. [PMID: 23391594 PMCID: PMC3936018 DOI: 10.1016/j.brainres.2013.01.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 01/12/2013] [Indexed: 02/08/2023]
Abstract
Understanding the cognitive impact of endogenously derived, and exogenously administered, hormone alterations is necessary for developing hormone treatments to support healthy brain function in women, especially during aging. The increasing number of studies in the burgeoning area of translational cognitive neuroendocrinology has revealed numerous factors that influence the extent and direction of female steroid effects on cognition. Here, we discuss the decision processes underlying the design of rodent hormone manipulation experiments evaluating learning and memory. It is noted that even when beginning with a clear hypothesis-driven question, there are numerous factors to consider in order to solidify a sound experimental design that will yield clean, interpretable results. Decisions and considerations include: age of animals at hormone administration and test, ovariectomy implementation, when to administer hormones relative to ovarian hormone loss, how and whether to monitor the estrous cycle if animals are ovary-intact, dose of hormone, administration route of hormone, hormone treatment confirmation protocols, handling procedures required for hormone administration and treatment confirmation, cognitive domains to be tested and which mazes should be utilized to test these cognitive domains, and control measures to be used. A balanced view of optimal design and realistic experimental practice and protocol is presented. The emerging results from translational cognitive neuroendocrinology studies have been diverse, but also enlightening and exciting as we realize the broad scope and powerful nature of ovarian hormone effects on the brain and its function. We must design, implement, and interpret hormone and cognition experiments with sensitivity to these tenets, acknowledging and respecting the breadth and depth of the impact gonadal hormones have on brain functioning and its rich plasticity. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- S.E. Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer’s Consortium, USA
| | - H.A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer’s Consortium, USA
| |
Collapse
|
46
|
Twining RC, Tuscher JJ, Doncheck EM, Frick KM, Mueller D. 17β-estradiol is necessary for extinction of cocaine seeking in female rats. Learn Mem 2013; 20:300-6. [PMID: 23676202 DOI: 10.1101/lm.030304.113] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Human and preclinical models of addiction demonstrate that gonadal hormones modulate acquisition of drug seeking. Little is known, however, about the effects of these hormones on extinction of drug-seeking behavior. Here, we investigated how 17β-estradiol (E₂) affects expression and extinction of cocaine seeking in female rats. Using a conditioned place preference (CPP) paradigm, ovariectomized rats were maintained throughout conditioning with 2 d of E₂ treatment followed by 2 d of vehicle treatment, or were injected with E₂ daily. Hormone injections were paired or explicitly unpaired with place conditioning sessions. Expression of a cocaine CPP was of equal magnitude regardless of conditioning protocol, suggesting that E₂ levels during conditioning did not affect subsequent CPP expression. During extinction, daily E₂ administration initially enhanced expression of the cocaine CPP, but resulted in significantly faster extinction compared to controls. Whereas E₂-treated rats were extinguished within 8 d, vehicle-treated rats maintained CPP expression for more than a month, indicative of perseveration. To determine whether E₂ could rescue extinction in these rats, half were given daily E₂ treatment and half were given vehicle. E₂-treated rats showed rapid extinction, whereas vehicle-treated rats continued to perseverate. These data demonstrate for the first time that E₂ is necessary for extinction of cocaine seeking in female rats, and that it promotes rapid extinction when administered daily. Clinically, these findings suggest that monitoring and maintaining optimal E₂ levels during exposure therapy would facilitate therapeutic interventions for female cocaine addicts.
Collapse
Affiliation(s)
- Robert C Twining
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | | | | | | | | |
Collapse
|
47
|
Remage-Healey L. Brain estrogen signaling effects acute modulation of acoustic communication behaviors: A working hypothesis. Bioessays 2012; 34:1009-16. [PMID: 23065844 DOI: 10.1002/bies.201200081] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Although estrogens are widely considered circulating "sex steroid hormones" typically associated with female reproduction, recent evidence suggests that estrogens can act as local modulators of brain circuits in both males and females. The functional implications of this newly characterized estrogen signaling system have begun to emerge. This essay summarizes evidence in support of the hypothesis that the rapid production of estrogens in brain circuits can drive acute changes in both the production and perception of acoustic communication behaviors. These studies have revealed two fundamental neurobiological concepts: (1) estrogens can be locally produced in brain circuits, independent of levels in nearby circuits and in the circulation and (2) estrogens can have very rapid effects within these brain circuits to modulate social vocalizations, acoustic processing, and sensorimotor integration. This vertebrate-wide span of research, including vocalizing fishes, amphibians, and birds, emphasizes the importance of comparative model systems in understanding principles of neurobiology.
Collapse
Affiliation(s)
- Luke Remage-Healey
- Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
48
|
Changing neuroestrogens within the auditory forebrain rapidly transform stimulus selectivity in a downstream sensorimotor nucleus. J Neurosci 2012; 32:8231-41. [PMID: 22699904 DOI: 10.1523/jneurosci.1114-12.2012] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The activity of sensory circuits is shaped by neuromodulators, which can have downstream consequences for both sensorimotor integration and behavioral output. Recent evidence indicates that brain-derived estrogens ("neuroestrogens") can act as local circuit modulators in the songbird auditory forebrain. Specifically, neuroestrogens fluctuate in the auditory caudomedial nidopallium (NCM) during social interactions and in response to song stimuli. Within minutes of elevation, neuroestrogens also enhance auditory response properties of NCM neurons, and acute blockade of estrogen production in NCM disrupts behavioral song preferences. Here, we test the hypothesis that fluctuating neuroestrogens within NCM influence stimulus selectivity in a downstream sensorimotor nucleus (HVC, used as a proper name) that receives indirect auditory input from NCM. Dual extracellular recordings coupled with retrodialysis delivery show that song selectivity in HVC is rapidly enhanced by increasing neuroestrogens in NCM in adult males. Conversely, inhibiting neuroestrogen production in NCM causes a rapid decline in song selectivity in HVC, demonstrating the endogenous nature of this modulatory network. In contrast, HVC selectivity is unaffected by neuroestrogen delivery to either nearby caudomedial mesopallium or into HVC itself, indicating that neuroestrogen actions are restricted to NCM. In juvenile males, identical neuroestrogen treatment in NCM also does not alter HVC selectivity, consistent with a developmental maturation of the auditory network. Lastly, the rapid actions of estrogens leading to enhanced HVC selectivity appear to be mediated by membrane-bound receptors in NCM. These findings indicate that steroid-dependent modulation of sensory processing is not locally restricted and can be transmitted transynaptically to influence downstream sensorimotor and premotor targets.
Collapse
|