1
|
Copenhaver AE, LeGates TA. Sex-Specific Mechanisms Underlie Long-Term Potentiation at Hippocampus→Medium Spiny Neuron Synapses in the Medial Shell of the Nucleus Accumbens. J Neurosci 2024; 44:e0100242024. [PMID: 38806250 PMCID: PMC11223474 DOI: 10.1523/jneurosci.0100-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
Sex differences have complicated our understanding of the neurobiological basis of many behaviors that are key for survival. As such, continued elucidation of the similarities and differences between sexes is necessary to gain insight into brain function and vulnerability. The connection between the hippocampus (Hipp) and nucleus accumbens (NAc) is a crucial site where modulation of neuronal activity mediates reward-related behavior. Our previous work demonstrated that long-term potentiation (LTP) of Hipp→NAc synapses is rewarding, and mice can establish learned associations between LTP of these synapses and the contextual environment in which LTP occurred. Here, we investigated sex differences in the mechanisms underlying Hipp→NAc LTP using whole-cell electrophysiology and pharmacology. We observed similarities in basal synaptic strength between males and females and found that LTP occurs postsynaptically with similar magnitudes in both sexes. However, key sex differences emerged as LTP in males required NMDA receptors (NMDAR), whereas LTP in females utilized an NMDAR-independent mechanism involving L-type voltage-gated Ca2+ channels (VGCCs) and estrogen receptor α (ERα). We also uncovered sex-similar features as LTP in both sexes depended on CaMKII activity and occurred independently of dopamine-1 receptor (D1R) activation. Our results have elucidated sex-specific molecular mechanisms for LTP in an integral pathway that mediates reward-related behaviors, emphasizing the importance of considering sex as a variable in mechanistic studies. Continued characterization of sex-specific mechanisms underlying plasticity will offer novel insight into the neurophysiological basis of behavior, with significant implications for understanding how diverse processes mediate behavior and contribute to vulnerability to developing psychiatric disorders.
Collapse
Affiliation(s)
- Ashley E Copenhaver
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
| | - Tara A LeGates
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| |
Collapse
|
2
|
Ichipi-Ifukor PC, Asagba SO, Achuba FI. Co-exposure to Aluminium and Cadmium Mediates Postpartum Maternal Variation in Brain Architecture and Behaviour of Mice; Involvement of Oxido-nitrergic and Cholinergic Mechanisms : Postpartum effects of Aluminium and Cadmium co-exposure in pregnancy. Biol Trace Elem Res 2024:10.1007/s12011-024-04218-0. [PMID: 38777875 DOI: 10.1007/s12011-024-04218-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Most research has not been done on the possible relationship between pregnant women's cross-metal exposures and postpartum neuroendocrine functions. The purpose of this study was to look into how co-exposure to aluminium chloride (AlCl3) and cadmium chloride (CdCl2) affected the neuroendocrine and neurometabolic changes in postpartum mice. A total of 24 adult pregnant female mice were used for the study. Group 1 served as control and received neither AlCl3 nor CdCl2 (n=6), group 2 comprised pregnant mice treated with AlCl3 (10mg/kg), group 3 with CdCl2 (1.5mg/kg), group 4 with a combination of AlCl3 (10 mg/kg) and CdCl2 (1.5 mg/kg).Oral treatment of animals was done daily from gestation day 7 to gestation day 20. Upon delivery and weaning on postnatal day 21 (PND 21), behavioural assessment was done on the postpartum mice and immediately followed by sacrifice for assessment of histological and neuroendocrine markers. Our findings revealed that the brain-to-body weight ratio was affected and brain oxidative stress was elevated in mice exposed to AlCl3 and CdCl2 during pregnancy. Given the strong association between postpartum hyperactivity, social interaction index, brain catalase and acetylcholinesterase activity, and the brain/body weight ratio, it is plausible that these effects have played a role in the adverse behavioural abnormalities observed in the postpartum maternal mice. Moreover, it was noted that in certain situations, co-exposures to the metals tended to have opposite effects to single metal exposures.
Collapse
|
3
|
Joue G, Navarro-Schröder T, Achtzehn J, Moffat S, Hennies N, Fuß J, Döller C, Wolbers T, Sommer T. Effects of estrogen on spatial navigation and memory. Psychopharmacology (Berl) 2024; 241:1037-1063. [PMID: 38407638 PMCID: PMC11031496 DOI: 10.1007/s00213-024-06539-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024]
Abstract
RATIONALE Animal studies suggest that the so-called "female" hormone estrogen enhances spatial navigation and memory. This contradicts the observation that males generally out-perform females in spatial navigation and tasks involving spatial memory. A closer look at the vast number of studies actually reveals that performance differences are not so clear. OBJECTIVES To help clarify the unclear performance differences between men and women and the role of estrogen, we attempted to isolate organizational from activational effects of estrogen on spatial navigation and memory. METHODS In a double-blind, placebo-controlled study, we tested the effects of orally administered estradiol valerate (E2V) in healthy, young women in their low-hormone menstrual cycle phase, compared to healthy, young men. Participants performed several first-person, environmentally rich, 3-D computer games inspired by spatial navigation and memory paradigms in animal research. RESULTS We found navigation behavior suggesting that sex effects dominated any E2 effects with men performing better with allocentric strategies and women with egocentric strategies. Increased E2 levels did not lead to general improvements in spatial ability in either sex but to behavioral changes reflecting navigation flexibility. CONCLUSION Estrogen-driven differences in spatial cognition might be better characterized on a spectrum of navigation flexibility rather than by categorical performance measures or skills.
Collapse
Affiliation(s)
- Gina Joue
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Tobias Navarro-Schröder
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030, Trondheim, Norway
| | - Johannes Achtzehn
- Department of Neurology with Experimental Neurology (CVK), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Scott Moffat
- School of Psychology, Georgia Institute of Technology, 654 Cherry Street, Atlanta, GA, 30332, USA
| | - Nora Hennies
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Johannes Fuß
- Institute of Forensic Psychiatry and Sex Research, University Duisburg-Essen, Hohlweg 26, 45147, Essen, Germany
| | - Christian Döller
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030, Trondheim, Norway
- Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1a, 04103, Leipzig, Germany
| | - Thomas Wolbers
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Tobias Sommer
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| |
Collapse
|
4
|
Been LE, Halliday AR, Blossom SM, Bien EM, Bernhard AG, Roth GE, Domenech Rosario KI, Pollock KB, Abramenko PE, Behbehani LM, Pascal GJ, Kelly ME. Long-Term Oral Tamoxifen Administration Decreases Brain-Derived Neurotrophic Factor in the Hippocampus of Female Long-Evans Rats. Cancers (Basel) 2024; 16:1373. [PMID: 38611051 PMCID: PMC11010888 DOI: 10.3390/cancers16071373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/24/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Tamoxifen, a selective estrogen receptor modulator (SERM), is commonly used as an adjuvant drug therapy for estrogen-receptor-positive breast cancers. Though effective at reducing the rate of cancer recurrence, patients often report unwanted cognitive and affective side effects. Despite this, the impacts of chronic tamoxifen exposure on the brain are poorly understood, and rodent models of tamoxifen exposure do not replicate the chronic oral administration seen in patients. We, therefore, used long-term ad lib consumption of medicated food pellets to model chronic tamoxifen exposure in a clinically relevant way. Adult female Long-Evans Hooded rats consumed tamoxifen-medicated food pellets for approximately 12 weeks, while control animals received standard chow. At the conclusion of the experiment, blood and brain samples were collected for analyses. Blood tamoxifen levels were measured using a novel ultra-performance liquid chromatography-tandem mass spectrometry assay, which found that this administration paradigm produced serum levels of tamoxifen similar to those in human patients. In the brain, brain-derived neurotrophic factor (BDNF) was visualized in the hippocampus using immunohistochemistry. Chronic oral tamoxifen treatment resulted in a decrease in BDNF expression across several regions of the hippocampus. These findings provide a novel method of modeling and measuring chronic oral tamoxifen exposure and suggest a putative mechanism by which tamoxifen may cause cognitive and behavioral changes reported by patients.
Collapse
Affiliation(s)
- Laura E. Been
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Amanda R. Halliday
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Sarah M. Blossom
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Elena M. Bien
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Anya G. Bernhard
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Grayson E. Roth
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Karina I. Domenech Rosario
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Karlie B. Pollock
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Petra E. Abramenko
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Leily M. Behbehani
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Gabriel J. Pascal
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
| | - Mary Ellen Kelly
- Department of Psychology and Neuroscience Program, Haverford College, Haverford, PA 19041, USA; (A.R.H.); (S.M.B.); (E.M.B.); (A.G.B.); (G.E.R.); (K.I.D.R.); (K.B.P.); (P.E.A.); (L.M.B.); (G.J.P.); (M.E.K.)
- Neuroscience Program, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Le AA, Palmer LC, Chavez J, Gall CM, Lynch G. Sex differences in the context dependency of episodic memory. Front Behav Neurosci 2024; 18:1349053. [PMID: 38516050 PMCID: PMC10956361 DOI: 10.3389/fnbeh.2024.1349053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Context contributes to multiple aspects of human episodic memory including segmentation and retrieval. The present studies tested if, in adult male and female mice, context influences the encoding of odors encountered in a single unsupervised sampling session of the type used for the routine acquisition of episodic memories. The three paradigms used differed in complexity (single vs. multiple odor cues) and period from sampling to testing. Results show that males consistently encode odors in a context-dependent manner: the mice discriminated novel from previously sampled cues when tested in the chamber of initial cue sampling but not in a distinct yet familiar chamber. This was independent of the interval between cue encounters or the latency from initial sampling to testing. In contrast, female mice acquired both single cues and the elements of multi-cue episodes, but recall of that information was dependent upon the surrounding context only when the cues were presented serially. These results extend the list of episodic memory features expressed by rodents and also introduce a striking and unexpected sex difference in context effects.
Collapse
Affiliation(s)
- Aliza A. Le
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
| | - Linda C. Palmer
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
| | - Jasmine Chavez
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
| | - Christine M. Gall
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - Gary Lynch
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
6
|
Wang X, Kong F, Liu Y, Lv S, Zhang K, Sun S, Liu J, Wang M, Cai X, Jin H, Yan S, Luo J. 17β-estradiol biosensors based on different bioreceptors and their applications. Front Bioeng Biotechnol 2024; 12:1347625. [PMID: 38357703 PMCID: PMC10864596 DOI: 10.3389/fbioe.2024.1347625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
17β-Estradiol (E2) is a critical sex steroid hormone, which has significant effects on the endocrine systems of both humans and animals. E2 is also believed to play neurotrophic and neuroprotective roles in the brain. Biosensors present a powerful tool to detect E2 because of their small, efficient, and flexible design. Furthermore, Biosensors can quickly and accurately obtain detection results with only a small sampling amount, which greatly meets the detection of the environment, food safety, medicine safety, and human body. This review focuses on previous studies of biosensors for detecting E2 and divides them into non-biometric sensors, enzyme biosensors, antibody biosensors, and aptamer biosensors according to different bioreceptors. The advantages, disadvantages, and design points of various bioreceptors for E2 detection are analyzed and summarized. Additionally, applications of different bioreceptors of E2 detection are presented and highlight the field of environmental monitoring, food and medicine safety, and disease detection in recent years. Finally, the development of E2 detection by biosensor is prospected.
Collapse
Affiliation(s)
- Xinyi Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Fanli Kong
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yaoyao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Shiya Lv
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Kui Zhang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Shutong Sun
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Mixia Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Hongyan Jin
- Obstetrics and Gynecology Department, Peking University First Hospital, Beijing, China
| | - Shi Yan
- Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
7
|
Copenhaver AE, LeGates TA. Sex-specific mechanisms underlie long-term potentiation at hippocampus-nucleus accumbens synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575709. [PMID: 38293132 PMCID: PMC10827060 DOI: 10.1101/2024.01.15.575709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Sex differences have complicated our understanding of the neurobiological basis of many behaviors that are key for survival. As such, continued elucidation of the similarities and differences between sexes is necessary in order to gain insight into brain function and vulnerability. The connection between the hippocampus (Hipp) and nucleus accumbens (NAc) is a crucial site where modulation of neuronal activity mediates reward-related behavior. Our previous work demonstrated that long-term potentiation (LTP) of Hipp-NAc synapses is rewarding, and that mice can make learned associations between LTP of these synapses and the contextual environment in which LTP occurred. Here, we investigate sex differences in the mechanisms underlying Hipp-NAc LTP using whole-cell electrophysiology and pharmacology. We found that males and females display similar magnitudes of Hipp-NAc LTP which occurs postsynaptically. However, LTP in females requires L-type voltage-gated Ca 2+ channels (VGCC) for postsynaptic Ca 2+ influx, while males rely on NMDA receptors (NMDAR). Additionally, females require estrogen receptor α (ERα) activity for LTP while males do not. These differential mechanisms converge as LTP in both sexes depends on CAMKII activity and occurs independently of dopamine-1 receptor (D1R) activation. Our results have elucidated sex-specific molecular mechanisms for LTP in an integral excitatory pathway that mediates reward-related behaviors, emphasizing the importance of considering sex as a variable in mechanistic studies. Continued characterization of sex-specific mechanisms underlying plasticity will offer novel insight into the neurophysiological basis of behavior, with significant implications for understanding how diverse processes mediate behavior and contribute to vulnerability to developing psychiatric disorders. SIGNIFICANCE STATEMENT Strengthening of Hipp-NAc synapses drives reward-related behaviors. Male and female mice have similar magnitudes of long-term potentiation (LTP) and both sexes have a predicted postsynaptic locus of plasticity. Despite these similarities, we illustrate here that sex-specific molecular mechanisms are used to elicit LTP. Given the bidirectional relationship between Hipp-NAc synaptic strength in mediating reward-related behaviors, the use of distinct molecular mechanisms may explain sex differences observed in stress susceptibility or response to rewarding stimuli. Discovery and characterization of convergent sex differences provides mechanistic insight into the sex-specific function of Hipp-NAc circuitry and has widespread implications for circuits mediating learning and reward-related behavior.
Collapse
|
8
|
Perović M, Heffernan EM, Einstein G, Mack ML. Learning exceptions to category rules varies across the menstrual cycle. Sci Rep 2023; 13:21999. [PMID: 38081874 PMCID: PMC10713535 DOI: 10.1038/s41598-023-48628-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Ways in which ovarian hormones affect cognition have been long overlooked despite strong evidence of their effects on the brain. To address this gap, we study performance on a rule-plus-exception category learning task, a complex task that requires careful coordination of core cognitive mechanisms, across the menstrual cycle (N = 171). Results show that the menstrual cycle distinctly affects exception learning in a manner that parallels the typical rise and fall of estradiol across the cycle. Participants in their high estradiol phase outperform participants in their low estradiol phase and demonstrate more rapid learning of exceptions than a male comparison group. A likely mechanism underlying this effect is estradiol's impact on pattern separation and completion pathways in the hippocampus. These results provide novel evidence for the effects of the menstrual cycle on category learning, and underscore the importance of considering female sex-related variables in cognitive neuroscience research.
Collapse
Affiliation(s)
- Mateja Perović
- Department of Psychology, University of Toronto, 100 St. George St., Toronto, ON, M5S 3J3, Canada.
| | - Emily M Heffernan
- Department of Psychology, University of Toronto, 100 St. George St., Toronto, ON, M5S 3J3, Canada
| | - Gillian Einstein
- Department of Psychology, University of Toronto, 100 St. George St., Toronto, ON, M5S 3J3, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Tema Genus, Linköping University, Linköping, Sweden
- Rotman Research Institute, Baycrest Hospital, Toronto, Canada
| | - Michael L Mack
- Department of Psychology, University of Toronto, 100 St. George St., Toronto, ON, M5S 3J3, Canada
| |
Collapse
|
9
|
Boyd HM, Frick KM, Kwapis JL. Connecting the Dots: Potential Interactions Between Sex Hormones and the Circadian System During Memory Consolidation. J Biol Rhythms 2023; 38:537-555. [PMID: 37464775 PMCID: PMC10615791 DOI: 10.1177/07487304231184761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Both the circadian clock and sex hormone signaling can strongly influence brain function, yet little is known about how these 2 powerful modulatory systems might interact during complex neural processes like memory consolidation. Individually, the molecular components and action of each of these systems have been fairly well-characterized, but there is a fundamental lack of information about how these systems cooperate. In the circadian system, clock genes function as timekeeping molecules that convey time-of-day information on a well-stereotyped cycle that is governed by the suprachiasmatic nucleus. Keeping time is particularly important to synchronize various physiological processes across the brain and body, including those that regulate memory consolidation. Similarly, sex hormones are powerful modulators of memory, with androgens, estrogens, and progestins, all influencing memory consolidation within memory-relevant brain regions like the hippocampus. Despite clear evidence that each system can influence memory individually, exactly how the circadian and hormonal systems might interact to impact memory consolidation remains unclear. Research investigating either sex hormone action or circadian gene function within memory-relevant brain regions has unveiled several notable places in which the two systems could interact to control memory. Here, we bring attention to known interactions between the circadian clock and sex hormone signaling. We then review sex hormone-mediated control of memory consolidation, highlighting potential nodes through which the circadian system might interact during memory formation. We suggest that the bidirectional relationship between these two systems is essential for proper control of memory formation based on an animal's hormonal and circadian state.
Collapse
Affiliation(s)
- Hannah M. Boyd
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| | - Karyn M. Frick
- Department of Psychology, University of Wisconsin–Milwaukee, Milwaukee, Wisconsin
| | - Janine L. Kwapis
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
10
|
MenezesdaSilva GM, Veiga ECDA, Simões MJ, Simões RS, Shiroma ME, Baracat MCP, Cavalcanti GS, Junior JMS, Baracat EC. Effects of estrogen and raloxifene on synaptic density in the hippocampal CA1 region of ovariectomized rats. Clinics (Sao Paulo) 2023; 78:100312. [PMID: 38016196 PMCID: PMC10698248 DOI: 10.1016/j.clinsp.2023.100312] [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: 07/24/2023] [Revised: 10/11/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023] Open
Abstract
INTRODUCTION The CA1 region of the hippocampus has an important role in learning and memory. It has been shown that estrogen deficiency may reduce the synaptic density in the region and that hormone replacement therapy may attenuate the reduction. OBJECTIVES This study aimed to evaluate the effects of estrogen and raloxifene on the synaptic density profile in the CA1 region of the hippocampus in ovariectomized rats. METHODS Sixty ovariectomized three-month-old virgin rats were randomized into six groups (n = 10). Treatments started either three days (early treatment) or sixty days (late treatment) after ovariectomy. The groups received propylene glycol vehicle (0.5 mL/animal/day), equine conjugated estrogens (50 μg/animal/day), or raloxifene (3 mg/kg/day) either early or late after ovariectomy. The drugs were administered orally by gavage for 30 days. At the end of the treatments, the animals were anesthetized and transcardially perfused with ether and saline solution. The brains were removed and prepared for analysis under transmission electron microscopy and later fixed. RESULTS Results showed a significant increase in the synaptic density profile of the hippocampal CA1 region in both the early estrogen (0.534 ± 0.026 µ/m2) and the early raloxifene (0.437 ± 0.012 µ/m2) treatment groups compared to the early or late vehicle-treated control groups (0.338 ± 0.038 µ/m2 and 0.277 ± 0.015 µ/m2 respectively). CONCLUSIONS The present data suggest that the raloxifene effect may be lower than that of estrogen, even early or late treatment, on synaptic density in the hippocampus.
Collapse
Affiliation(s)
- Glaucia Mara MenezesdaSilva
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Eduardo Carvalho de Arruda Veiga
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil.
| | - Manuel Jesus Simões
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil; Disciplina de Histologia e Biologia Celular do Departamento de Morfologia da Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Ricardo Santos Simões
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Marcos Eiji Shiroma
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Maria Cândida Pinheiro Baracat
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Givanna Santos Cavalcanti
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Jose Maria Soares Junior
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Edmund Chada Baracat
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| |
Collapse
|
11
|
Zuluaga MJ, Agrati D, Athaíde V, Ferreira A, Uriarte N. Fear response of rat pups to a non-aversive social stimulus: Evidence for the involvement of memory processes. Dev Psychobiol 2023; 65:e22417. [PMID: 37860902 DOI: 10.1002/dev.22417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/20/2023] [Accepted: 08/10/2023] [Indexed: 10/21/2023]
Abstract
Learning processes in rats during early development are importantly mediated by the mother, which represents the primary source of environmental information. This study aimed to determine whether aversive early experiences can induce the expression of pups' fear responses toward a non-aversive stimulus as a consequence of a memory process. First, we determined pups' fear responses toward an anesthetized female after being exposed to this stimulus or an empty cage together with their mothers from Postnatal Day (PNDs) 1 to 4. Second, we evaluated if the administration of the protein synthesis inhibitor cycloheximide (CHX; 0.2 mg/kg, subcutaneously (sc).) disrupted the reconsolidation processes and abolished the fear response on PND 9. Only female pups previously exposed to the female intruder expressed fear responses toward an anesthetized female on PND 8. CHX administration to female pups immediately after exposure to an anesthetized female on PND 8 suppressed fear responses on PND 9, indicating that the fear expression was the result of a memory process, probably mediated by the mother. These findings demonstrated that early experiences can shape responses to social stimuli in a sex-dependent manner and emphasize the critical role of the mother in influencing fear learning in a social context.
Collapse
Affiliation(s)
- María José Zuluaga
- Biofisicoquímica, Departamento de Ciencias Biológicas, Centro Universitario Regional Litoral Norte-Sede Salto, Universidad de la República, Salto, Uruguay
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Daniella Agrati
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Vanessa Athaíde
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Annabel Ferreira
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Natalia Uriarte
- Laboratorio de Neurociencias, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
12
|
Arjmand S, Bender D, Jakobsen S, Wegener G, Landau AM. Peering into the Brain's Estrogen Receptors: PET Tracers for Visualization of Nuclear and Extranuclear Estrogen Receptors in Brain Disorders. Biomolecules 2023; 13:1405. [PMID: 37759805 PMCID: PMC10526964 DOI: 10.3390/biom13091405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Estrogen receptors (ERs) play a multitude of roles in brain function and are implicated in various brain disorders. The use of positron emission tomography (PET) tracers for the visualization of ERs' intricate landscape has shown promise in oncology but remains limited in the context of brain disorders. Despite recent progress in the identification and development of more selective ligands for various ERs subtypes, further optimization is necessary to enable the reliable and efficient imaging of these receptors. In this perspective, we briefly touch upon the significance of estrogen signaling in the brain and raise the setbacks associated with the development of PET tracers for identification of specific ERs subtypes in the brain. We then propose avenues for developing efficient PET tracers to non-invasively study the dynamics of ERs in the brain, as well as neuropsychiatric diseases associated with their malfunction in a longitudinal manner. This perspective puts several potential candidates on the table and highlights the unmet needs and areas requiring further research to unlock the full potential of PET tracers for ERs imaging, ultimately aiding in deepening our understanding of ERs and forging new avenues for potential therapeutic strategies.
Collapse
Affiliation(s)
- Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Dirk Bender
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Steen Jakobsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
| | - Anne M. Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| |
Collapse
|
13
|
Fleischer AW, Frick KM. New perspectives on sex differences in learning and memory. Trends Endocrinol Metab 2023; 34:526-538. [PMID: 37500421 PMCID: PMC10617789 DOI: 10.1016/j.tem.2023.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 07/29/2023]
Abstract
Females have historically been disregarded in memory research, including the thousands of studies examining roles for the hippocampus, medial prefrontal cortex, and amygdala in learning and memory. Even when included, females are often judged based on male-centric behavioral and neurobiological standards, generating and perpetuating scientific stereotypes that females exhibit worse memories compared with males in domains such as spatial navigation and fear. Recent research challenges these dogmas by identifying sex-specific strategies in common memory tasks. Here, we discuss rodent data illustrating sex differences in spatial and fear memory, as well as the neural mechanisms underlying memory formation. The influence of sex steroid hormones in both sexes is discussed, as is the importance to basic and translational neuroscience of studying sex differences.
Collapse
Affiliation(s)
- Aaron W Fleischer
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA.
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA.
| |
Collapse
|
14
|
Cortés YI, Cai J, Daviglus M, Gallo LC, Lamar M, Isasi CR, Perreira KM. Reproductive period duration and cognitive function in postmenopausal Latina women in the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). Maturitas 2023; 174:23-29. [PMID: 37224791 PMCID: PMC10832364 DOI: 10.1016/j.maturitas.2023.04.270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/01/2023] [Accepted: 04/30/2023] [Indexed: 05/26/2023]
Abstract
OBJECTIVES A shorter reproductive period, a marker of estrogen exposure, has been related to cognitive impairment among older (>65 years) non-Hispanic White women. We explored whether reproductive period duration, age at menarche, and age at menopause are related to cognitive function among postmenopausal Hispanic/Latina women. METHODS This cross-sectional analysis used baseline (Visit 1: 2008-2011) data from 3630 postmenopausal women in the Hispanic Community Health Study/Study of Latinos. Reproductive period duration, age at menarche, and age at menopause were assessed by self-report. Cognitive function variables included global cognition, verbal learning, memory, verbal fluency, and processing speed. Associations between each reproductive event and cognitive function were examined using multivariable linear and logistic regression analyses accounting for the complex survey design of the study and adjusting for socio-demographics, parity, and cardiovascular risk factors. We assessed whether associations differed by type of menopause (natural versus surgical) and hormone therapy use. RESULTS The study population was on average aged 59 years, with a mean reproductive period duration of 35 years. Older age at menopause and a longer reproductive period were related to higher verbal learning (β = 0.04, SE = 0.02; p < 0.05) and processing speed (ß = 0.16, SE = 0.04; p < 0.001); associations were more pronounced for women with natural menopause. Older age at menarche was associated with lower scores on the digit symbol substitution test (ß = -0.62, SE = 0.15; p < 0.0001). There were no associations with global cognition. CONCLUSION Among postmenopausal Hispanic/Latinas, a longer reproductive period was related to more favorable cognitive measures of verbal learning and processing speed. Our findings support the hypothesis that greater lifetime exposure to estrogens may be associated with higher cognitive performance.
Collapse
Affiliation(s)
- Yamnia I Cortés
- University of North Carolina at Chapel Hill School of Nursing, Carrington Hall Campus Box 7460, Chapel Hill, NC 27599, USA.
| | - Jianwen Cai
- University of North Carolina at Chapel Hill Gillings School of Public Health, 135 Dauer Drive, Chapel Hill, NC 27599, USA.
| | - Martha Daviglus
- Institute for Minority Health Research, The University of Illinois College of Medicine, 1737 West Polk Street, Chicago, IL, USA.
| | - Linda C Gallo
- Department of Psychology and South Bay Latino Research Center, San Diego State University, San Diego, CA, USA.
| | - Melissa Lamar
- Institute for Minority Health Research, The University of Illinois College of Medicine, 1737 West Polk Street, Chicago, IL, USA; Department of Psychiatry & Behavioral Sciences and Rush Alzheimer's Disease Center, Rush University Medical Center, 17590 W Harrison Street, Chicago, IL, USA.
| | - Carmen R Isasi
- Albert Einstein College of Medicine, Department of Epidemiology & Population Health, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | - Krista M Perreira
- Department of Social Medicine, University of North Carolina at Chapel Hill, MacNider Hall Campus Box, 724, Chapel Hill, NC, USA.
| |
Collapse
|
15
|
Ryakiotakis E, Fousfouka D, Stamatakis A. Maternal neglect alters reward-anticipatory behavior, social status stability, and reward circuit activation in adult male rats. Front Neurosci 2023; 17:1201345. [PMID: 37521688 PMCID: PMC10375725 DOI: 10.3389/fnins.2023.1201345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/15/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Adverse early life experiences affect neuronal growth and maturation of reward circuits that modify behavior under reward predicting conditions. Previous studies demonstrate that rats undergoing denial of expected reward in the form of maternal contact (DER-animal model of maternal neglect) during early post-natal life developed anhedonia, aggressive play-fight behaviors and aberrant prefrontal cortex structure and neurochemistry. Although many studies revealed social deficiency following early-life stress most reports focus on individual animal tasks. Thus, attention needs to be given on the social effects during group tasks in animals afflicted by early life adversity. Methods To investigate the potential impact of the DER experience on the manifestation of behavioral responses induced by natural rewards, we evaluated: 1) naïve adult male sexual preference and performance, and 2) anticipatory behavior during a group 2-phase food anticipation learning task composed of a context-dependent and a cue-dependent learning period. Results DER rats efficiently spent time in the vicinity of and initiated sexual intercourse with receptive females suggesting an intact sexual reward motivation and consummation. Interestingly, during the context-dependent phase of food anticipation training DER rats displayed a modified exploratory activity and lower overall reward-context association. Moreover, during the cue-dependent phase DER rats displayed a mild deficit in context-reward association while increased cue-dependent locomotion. Additionally, DER rats displayed unstable food access priority following food presentation. These abnormal behaviours were accompanied by overactivation of the ventral prefrontal cortex and nucleus accumbens, as assessed by pCREB levels. Conclusions/discussion Collectively, these data show that the neonatal DER experience resulted in adulthood in altered activation of the reward circuitry, interfered with the normal formation of context-reward associations, and disrupted normal reward access hierarchy formation. These findings provide additional evidence to the deleterious effects of early life adversity on reward system, social hierarchy formation, and brain function.
Collapse
Affiliation(s)
- Ermis Ryakiotakis
- Laboratory of Biology-Biochemistry, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra Fousfouka
- Laboratory of Biology-Biochemistry, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
- MSc Program in Molecular Biomedicine, Medical School of National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Stamatakis
- Laboratory of Biology-Biochemistry, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
16
|
Melchinger P, Garcia BM. Mitochondria are midfield players in steroid synthesis. Int J Biochem Cell Biol 2023; 160:106431. [PMID: 37207805 DOI: 10.1016/j.biocel.2023.106431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Steroids are important membrane components and signaling metabolites and thus are required for cellular homeostasis. All mammalian cells retain the ability to uptake and synthesize steroids. Dysregulation of steroid levels leads to profound effects on cellular function and organismal health. Hence it comes as no surprise that steroid synthesis is tightly regulated. It is well established that the main site for steroid synthesis and regulation is the endoplasmic reticulum. However, mitochondria are essential for: (1) cholesterol production (the precursor of all steroids) by exporting citrate and; (2) the products of steroidogenesis (such as mineralocorticoids and glucocorticoids). In this review, we describe the midfield player role of mitochondria in steroid synthesis and bring the idea of mitochondria actively participating in steroid synthesis regulation. A better understanding of the mitochondrial regulatory roles in steroid synthesis would open new avenues to targeted approaches aiming to control steroid levels.
Collapse
Affiliation(s)
- Philipp Melchinger
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Department of Biological Sciences, University of Cologne, Cologne, Germany.
| | | |
Collapse
|
17
|
Gall CM, Le AA, Lynch G. Sex differences in synaptic plasticity underlying learning. J Neurosci Res 2023; 101:764-782. [PMID: 33847004 PMCID: PMC10337639 DOI: 10.1002/jnr.24844] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 11/09/2022]
Abstract
Although sex differences in learning behaviors are well documented, sexual dimorphism in the synaptic processes of encoding is only recently appreciated. Studies in male rodents have built upon the discovery of long-term potentiation (LTP), and acceptance of this activity-dependent increase in synaptic strength as a mechanism of encoding, to identify synaptic receptors and signaling activities that coordinate the activity-dependent remodeling of the subsynaptic actin cytoskeleton that is critical for enduring potentiation and memory. These molecular substrates together with other features of LTP, as characterized in males, have provided an explanation for a range of memory phenomena including multiple stages of consolidation, the efficacy of spaced training, and the location of engrams at the level of individual synapses. In the present report, we summarize these findings and describe more recent results from our laboratories showing that in females the same actin regulatory mechanisms are required for hippocampal LTP and memory but, in females only, the engagement of both modulatory receptors such as TrkB and synaptic signaling intermediaries including Src and ERK1/2 requires neuron-derived estrogen and signaling through membrane-associated estrogen receptor α (ERα). Moreover, in association with the additional ERα involvement, females exhibit a higher threshold for hippocampal LTP and spatial learning. We propose that the distinct LTP threshold in females contributes to as yet unappreciated sex differences in information processing and features of learning and memory.
Collapse
Affiliation(s)
- Christine M. Gall
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Aliza A. Le
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Gary Lynch
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| |
Collapse
|
18
|
Mulvey B, Frye HE, Lintz T, Fass S, Tycksen E, Nelson EC, Morón JA, Dougherty JD. Cnih3 Deletion Dysregulates Dorsal Hippocampal Transcription across the Estrous Cycle. eNeuro 2023; 10:ENEURO.0153-22.2023. [PMID: 36849260 PMCID: PMC10027183 DOI: 10.1523/eneuro.0153-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 03/01/2023] Open
Abstract
In females, the hippocampus, a critical brain region for coordination of learning, memory, and behavior, displays altered physiology and behavioral output across the estrous or menstrual cycle. However, the molecular effectors and cell types underlying these observed cyclic changes have only been partially characterized to date. Recently, profiling of mice null for the AMPA receptor trafficking gene Cnih3 have demonstrated estrous-dependent phenotypes in dorsal hippocampal synaptic plasticity, composition, and learning/memory. We therefore profiled dorsal hippocampal transcriptomes from female mice in each estrous cycle stage, and contrasted it with that of males, across wild-type (WT) and Cnih3 mutants. In wild types, we identified only subtle differences in gene expression between the sexes, while comparing estrous stages to one another revealed up to >1000 differentially expressed genes (DEGs). These estrous-responsive genes are especially enriched in gene markers of oligodendrocytes and the dentate gyrus, and in functional gene sets relating to estrogen response, potassium channels, and synaptic gene splicing. Surprisingly, Cnih3 knock-outs (KOs) showed far broader transcriptomic differences between estrous cycle stages and males. Moreover, Cnih3 knock-out drove subtle but extensive expression changes accentuating sex differential expression at diestrus and estrus. Altogether, our profiling highlights cell types and molecular systems potentially impacted by estrous-specific gene expression patterns in the adult dorsal hippocampus, enabling mechanistic hypothesis generation for future studies of sex-differential neuropsychiatric function and dysfunction. Moreover, these findings suggest an unrecognized role of Cnih3 in buffering against transcriptional effects of estrous, providing a candidate molecular mechanism to explain estrous-dependent phenotypes observed with Cnih3 loss.
Collapse
Affiliation(s)
- Bernard Mulvey
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Hannah E Frye
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Tania Lintz
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Stuart Fass
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Eric Tycksen
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110
| | - Elliot C Nelson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Jose A Morón
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
19
|
Potier M, Maitre M, Leste-Lasserre T, Marsicano G, Chaouloff F, Marighetto A. Age-dependent effects of estradiol on temporal memory: A role for the type 1 cannabinoid receptor? Psychoneuroendocrinology 2023; 148:106002. [PMID: 36521252 DOI: 10.1016/j.psyneuen.2022.106002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
This study investigated in male mice how age modulates the effects of acute 17β-estradiol (E2) on dorsal CA1 (dCA1)-dependent retention of temporal associations, which are critical for declarative memory. E2 was systemically injected to young (3-4 months old) and aged (22-24 months old) adult mice either (i) 1 h before the acquisition of an auditory trace fear conditioning (TFC) procedure allowing the assessment of temporal memory retention 24 h later or (ii) during in vivo electrophysiological recordings of CA3 to dCA1 synaptic efficacy under anesthesia. In young mice, E2 induced parallel dose-dependent reductions in memory and synaptic efficacy, i.e. an impairment in TFC retention and a long-term (NMDA receptor-dependent) depression of dCA1 synaptic efficacy as assessed by field excitatory postsynaptic potentials. In contrast, E2 tended to improved TFC retention whilst failing to change synaptic efficacy in aged mice. Age-dependent effects of E2 treatment were confirmed by immunohistochemical analyses of TFC acquisition-elicited dCA1 Fos activation. Thus, such an activation was respectively reduced and enhanced in young and aged E2-treated mice, compared to vehicle treatments. Hippocampal mRNA expression of estrogen receptors by RT-PCR analyses revealed an age-related increase in each receptor mRNA expression. In keeping with the key role of the endocannabinoid system in memory processes and CA3 to dCA1 synaptic plasticity, we next examined the role of cannabinoid type 1 receptors (CB1-R) in the aforementioned age-dependent effects of E2. Having confirmed that mRNA expression of CB1-R diminishes with age, we then observed that the deleterious effects of E2 on both memory and synaptic efficacy were both prevented by the CB1-R antagonist Rimonabant whilst being absent in CB1-R knock out mice. This study (i) reveals age-dependent effects of acute E2 on temporal memory and CA3 to dCA1 synaptic efficacy and (ii) suggests a key role of CB1-R in mediating E2 deleterious effects in young adulthood. Aging-related reductions in CB1-R might thus underlie E2 paradoxical effects across age.
Collapse
Affiliation(s)
- Mylène Potier
- Pathophysiology of Declarative Memory, INSERM U1215, Neurocentre Magendie, Bordeaux, France; University of Bordeaux, Bordeaux, France.
| | - Marlène Maitre
- PUMA, INSERM U1215, Neurocentre Magendie, Bordeaux, France
| | | | - Giovanni Marsicano
- Endocannabinoids & NeuroAdaptation, INSERM U1215, Neurocentre Magendie, Bordeaux, France; University of Bordeaux, Bordeaux, France
| | - Francis Chaouloff
- Endocannabinoids & NeuroAdaptation, INSERM U1215, Neurocentre Magendie, Bordeaux, France; University of Bordeaux, Bordeaux, France.
| | - Aline Marighetto
- Pathophysiology of Declarative Memory, INSERM U1215, Neurocentre Magendie, Bordeaux, France; University of Bordeaux, Bordeaux, France
| |
Collapse
|
20
|
Rocks D, Kundakovic M. Hippocampus-based behavioral, structural, and molecular dynamics across the estrous cycle. J Neuroendocrinol 2023; 35:e13216. [PMID: 36580348 PMCID: PMC10050126 DOI: 10.1111/jne.13216] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/19/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022]
Abstract
The activity of neurons in the rodent hippocampus contributes to diverse behaviors, with the activity of ventral hippocampal neurons affecting behaviors related to anxiety and emotion regulation, and the activity of dorsal hippocampal neurons affecting performance in learning- and memory-related tasks. Hippocampal cells also express receptors for ovarian hormones, estrogen and progesterone, and are therefore affected by physiological fluctuations of those hormones that occur over the rodent estrous cycle. In this review, we discuss the effects of cycling ovarian hormones on hippocampal physiology. Starting with behavior, we explore the role of the estrous cycle in regulating hippocampus-dependent behaviors. We go on to detail the cellular mechanisms through which cycling estrogen and progesterone, through changes in the structural and functional properties of hippocampal neurons, may be eliciting these changes in behavior. Then, providing a basis for these cellular changes, we outline the epigenetic, chromatin regulatory mechanisms through which ovarian hormones, by binding to their receptors, can affect the regulation of behavior- and synaptic plasticity-related genes in hippocampal neurons. We also highlight an unconventional role that chromatin dynamics may have in regulating neuronal function across the estrous cycle, including in sex hormone-driven X chromosome plasticity and hormonally-induced epigenetic priming. Finally, we discuss directions for future studies and the translational value of the rodent estrous cycle for understanding the effects of the human menstrual cycle on hippocampal physiology and brain disease risk.
Collapse
Affiliation(s)
- Devin Rocks
- Department of Biological Sciences, Fordham University; Bronx, NY, USA
| | - Marija Kundakovic
- Department of Biological Sciences, Fordham University; Bronx, NY, USA
| |
Collapse
|
21
|
Klotho Regulated by Estrogen Plays a Key Role in Sex Differences in Stress Resilience in Rats. Int J Mol Sci 2023; 24:ijms24021206. [PMID: 36674721 PMCID: PMC9862442 DOI: 10.3390/ijms24021206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Klotho (KL) is a glycosyl hydrolase and aging-suppressor gene. Stress is a risk factor for depression and anxiety, which are highly comorbid with each other. The aim of this study is to determine whether KL is regulated by estrogen and plays an important role in sex differences in stress resilience. Our results showed that KL is regulated by estrogen in rat hippocampal neurons in vivo and in vitro and is essential for the estrogen-mediated increase in the number of presynaptic vesicular glutamate transporter 1 (Vglut1)-positive clusters on the dendrites of hippocampal neurons. The role of KL in sex differences in stress response was examined in rats using 3-week chronic unpredictable mild stress (CUMS). CUMS produced a deficit in spatial learning and memory, anhedonic-like behaviors, and anxiety-like behaviors in male but not female rats, which was accompanied by a reduction in KL protein levels in the hippocampus of male but not female rats. This demonstrated the resilience of female rats to CUMS. Interestingly, the knockdown of KL protein levels in the rat hippocampus of both sexes caused a decrease in stress resilience in both sexes, especially in female rats. These results suggest that the regulation of KL by estrogen plays an important role in estrogen-mediated synapse formation and that KL plays a critical role in the sex differences in cognitive deficit, anhedonic-like behaviors, and anxiety-like behaviors induced by chronic stress in rats, highlighting an important role of KL in sex differences in stress resilience.
Collapse
|
22
|
Linhares SSG, Meurer YDSR, de Aquino ACQ, Câmara DDA, Brandão LEM, Fiuza FP, Lima RH, Engelberth RCJG, Cavalcante JS. Prenatal exposure to fluoxetine modulates emotionality and aversive memory in male and female rat offspring. Behav Pharmacol 2022; 33:575-588. [PMID: 36256730 DOI: 10.1097/fbp.0000000000000705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During pregnancy, women are prone to depression, for which selective serotonin reuptake inhibitors (SSRIs), such as fluoxetine, are usually the first-line treatment. However, fluoxetine can cross the placental barrier and affect fetuses, causing changes in serotonin levels early in life. Long-term effects in the brain circuits that control cognitive and emotional behavior are related to early fluoxetine exposure during development. In this study, we aimed to investigate whether fluoxetine exposure (10 mg/kg/day) from the 13th gestational day (GD13) to GD21 may lead to behavioral emotional-cognitive changes in male and female rat offspring approximately 90 days postnatally (~PN90). We have analyzed the performance of individuals in the open field and in the plus-maze discriminative avoidance task, which assesses anxiety and learning/memory processing behaviors. We have found that prenatal (GD13-GD21) exposure to fluoxetine strengthened aversive memory and induced higher anxiety levels in males, and quick extinction of aversive memory in females. Taken together, these results suggest that early exposure to fluoxetine impairs the basal state of anxiety and the cognitive functions of rats during adulthood, which may be in a sex-specific manner because males appear more susceptible than females.
Collapse
Affiliation(s)
- Sarah Sophia G Linhares
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Ywlliane da Silva R Meurer
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Antônio Carlos Queiroz de Aquino
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Diego de Aquino Câmara
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Felipe Porto Fiuza
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Ramón Hypolito Lima
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Rovena Clara J G Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Jeferson Souza Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
23
|
Tsan L, Sun S, Hayes AMR, Bridi L, Chirala LS, Noble EE, Fodor AA, Kanoski SE. Early life Western diet-induced memory impairments and gut microbiome changes in female rats are long-lasting despite healthy dietary intervention. Nutr Neurosci 2022; 25:2490-2506. [PMID: 34565305 PMCID: PMC8957635 DOI: 10.1080/1028415x.2021.1980697] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Western diet consumption during adolescence results in hippocampus (HPC)-dependent memory impairments and gut microbiome dysbiosis. Whether these adverse outcomes persist in adulthood following healthy dietary intervention is unknown. Here we assessed the short- and long-term effects of adolescent consumption of a Western diet enriched with either sugar or both sugar and fat on metabolic outcomes, HPC function, and gut microbiota. METHODS Adolescent female rats (PN 26) were fed a standard chow diet (CHOW), chow with access to 11% sugar solution (SUG), or a junk food cafeteria-style diet (CAF) containing various foods high in fat and/or sugar. During adulthood (PN 65+), metabolic outcomes, HPC-dependent memory, and gut microbial populations were evaluated. In a subsequent experiment, these outcomes were evaluated following a 5-week dietary intervention where CAF and SUG groups were maintained on standard chow alone. RESULTS Both CAF and SUG groups demonstrated impaired HPC-dependent memory, increased adiposity, and altered gut microbial populations relative to the CHOW group. However, impaired peripheral glucose regulation was only observed in the SUG group. When examined following a healthy dietary intervention in a separate experiment, metabolic dysfunction was not observed in either the CAF or SUG group, whereas HPC-dependent memory impairments were observed in the CAF but not the SUG group. In both groups the composition of the gut microbiota remained distinct from CHOW rats after the dietary intervention. CONCLUSIONS While the metabolic impairments associated with adolescent junk food diet consumption are not present in adulthood following dietary intervention, the HPC-dependent memory impairments and the gut microbiome dysbiosis persist.
Collapse
Affiliation(s)
- Linda Tsan
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Shan Sun
- Department of Bioinformatics and Genomics at the University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Anna M. R. Hayes
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Lana Bridi
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Lekha S. Chirala
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Emily E. Noble
- Department of Foods and Nutrition, University of Georgia, Athens, GA, USA
| | - Anthony A. Fodor
- Department of Bioinformatics and Genomics at the University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Scott E. Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
24
|
Honig MG, Del Mar NA, Moore BM, Reiner A. Raloxifene Mitigates Emotional Deficits after Mild Traumatic Brain Injury in Mice. Neurotrauma Rep 2022; 3:534-544. [DOI: 10.1089/neur.2022.0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Marcia G. Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Nobel A. Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Bob M. Moore
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
25
|
Automated classification of estrous stage in rodents using deep learning. Sci Rep 2022; 12:17685. [PMID: 36271290 PMCID: PMC9587051 DOI: 10.1038/s41598-022-22392-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 10/13/2022] [Indexed: 01/18/2023] Open
Abstract
The rodent estrous cycle modulates a range of biological functions, from gene expression to behavior. The cycle is typically divided into four stages, each characterized by distinct hormone concentration profiles. Given the difficulty of repeatedly sampling plasma steroid hormones from rodents, the primary method for classifying estrous stage is by identifying vaginal epithelial cell types. However, manual classification of epithelial cell samples is time-intensive and variable, even amongst expert investigators. Here, we use a deep learning approach to achieve classification accuracy at expert level. Due to the heterogeneity and breadth of our input dataset, our deep learning approach ("EstrousNet") is highly generalizable across rodent species, stains, and subjects. The EstrousNet algorithm exploits the temporal dimension of the hormonal cycle by fitting classifications to an archetypal cycle, highlighting possible misclassifications and flagging anestrus phases (e.g., pseudopregnancy). EstrousNet allows for rapid estrous cycle staging, improving the ability of investigators to consider endocrine state in their rodent studies.
Collapse
|
26
|
Hokenson RE, Alam YH, Short AK, Jung S, Jang C, Baram TZ. Sex-dependent effects of multiple acute concurrent stresses on memory: a role for hippocampal estrogens. Front Behav Neurosci 2022; 16:984494. [PMID: 36160685 PMCID: PMC9492881 DOI: 10.3389/fnbeh.2022.984494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022] Open
Abstract
Memory disruption commonly follows chronic stress, whereas acute stressors are generally benign. However, acute traumas such as mass shootings or natural disasters—lasting minutes to hours and consisting of simultaneous physical, social, and emotional stresses—are increasingly recognized as significant risk factors for memory problems and PTSD. Our prior work has revealed that these complex stresses (concurrent multiple acute stresses: MAS) disrupt hippocampus-dependent memory in male rodents. In females, the impacts of MAS are estrous cycle-dependent: MAS impairs memory during early proestrus (high estrogens phase), whereas the memory of female mice stressed during estrus (low estrogens phase) is protected. Female memory impairments limited to high estrogens phases suggest that higher levels of estrogens are necessary for MAS to disrupt memory, supported by evidence that males have higher hippocampal estradiol than estrous females. To test the role of estrogens in stress-induced memory deficits, we blocked estrogen production using aromatase inhibitors. A week of blockade protected male and female mice from MAS-induced memory disturbances, suggesting that high levels of estrogens are required for stress-provoked memory impairments in both males and females. To directly quantify 17β-estradiol in murine hippocampus we employed both ELISA and mass spectrometry and identified significant confounders in both procedures. Taken together, the cross-cycle and aromatase studies in males and females support the role for high hippocampal estrogens in mediating the effect of complex acute stress on memory. Future studies focus on the receptors involved, the longevity of these effects, and their relation to PTSD-like behaviors in experimental models.
Collapse
Affiliation(s)
- Rachael E. Hokenson
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, United States
- *Correspondence: Rachael E. Hokenson
| | - Yasmine H. Alam
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Annabel K. Short
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA =, United States
| | - Sunhee Jung
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Tallie Z. Baram
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA =, United States
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
27
|
Saleki K, Banazadeh M, Saghazadeh A, Rezaei N. Aging, testosterone, and neuroplasticity: friend or foe? Rev Neurosci 2022; 34:247-273. [PMID: 36017670 DOI: 10.1515/revneuro-2022-0033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/03/2022] [Indexed: 11/15/2022]
Abstract
Neuroplasticity or neural plasticity implicates the adaptive potential of the brain in response to extrinsic and intrinsic stimuli. The concept has been utilized in different contexts such as injury and neurological disease. Neuroplasticity mechanisms have been classified into neuroregenerative and function-restoring processes. In the context of injury, neuroplasticity has been defined in three post-injury epochs. Testosterone plays a key yet double-edged role in the regulation of several neuroplasticity alterations. Research has shown that testosterone levels are affected by numerous factors such as age, stress, surgical procedures on gonads, and pharmacological treatments. There is an ongoing debate for testosterone replacement therapy (TRT) in aging men; however, TRT is more useful in young individuals with testosterone deficit and more specific subgroups with cognitive dysfunction. Therefore, it is important to pay early attention to testosterone profile and precisely uncover its harms and benefits. In the present review, we discuss the influence of environmental factors, aging, and gender on testosterone-associated alterations in neuroplasticity, as well as the two-sided actions of testosterone in the nervous system. Finally, we provide practical insights for further study of pharmacological treatments for hormonal disorders focusing on restoring neuroplasticity.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,USERN Office, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| | - Mohammad Banazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, 76169 13555 Kerman, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 14176 13151 Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| |
Collapse
|
28
|
Pinizzotto CC, Patwardhan A, Aldarondo D, Kritzer MF. Task-specific effects of biological sex and sex hormones on object recognition memories in a 6-hydroxydopamine-lesion model of Parkinson's disease in adult male and female rats. Horm Behav 2022; 144:105206. [PMID: 35653829 DOI: 10.1016/j.yhbeh.2022.105206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/19/2022] [Accepted: 05/21/2022] [Indexed: 01/13/2023]
Abstract
Many patients with Parkinson's disease (PD) experience cognitive or memory impairments with few therapeutic options available to mitigate them. This has fueled interest in determining how factors including sex and sex hormones modulate higher order function in this disease. The objective of this study was to use the Novel Object Recognition (NOR) and Object-in-Place (OiP) paradigms to compare the effects of a bilateral neostriatal 6-hydroxydopamine (6-OHDA) lesion model of PD in gonadally intact male and female rats, in orchidectomized male rats and in orchidectomized males supplemented with 17β-estradiol or testosterone propionate on measures of recognition memory similar to those at risk in PD. These studies showed that 6-ODHA lesions impaired discrimination in both tasks in males but not females. Further, 6-OHDA lesions disrupted NOR performance similarly in all males regardless of whether they were gonadally intact, orchidectomized or hormone-supplemented. In contrast, OiP performance was disrupted in males that were orchidectomized or 6-OHDA-lesioned but was spared in orchidectomized and orchidectomized, 6-OHDA lesioned males supplemented with 17β-estradiol. The distinct effects that sex and/or sex hormones have on 6-OHDA lesion-induced NOR vs. OiP deficits identified here also differ from corresponding impacts recently described for 6-OHDA lesion-induced deficits in spatial working memory and episodic memory. Together, the collective data provide strong evidence for effects of sex and sex hormones on cognition and memory in PD as being behavioral task and behavioral domain specific. This specificity could explain why a cohesive clinical picture of endocrine impacts on higher order function in PD has remained elusive.
Collapse
Affiliation(s)
- Claudia C Pinizzotto
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York 11794, USA.
| | - Aishwarya Patwardhan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York 11794, USA.
| | - Daniel Aldarondo
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York 11794, USA.
| | - Mary F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York 11794, USA.
| |
Collapse
|
29
|
Noel SC, Fortin-Hamel L, Haque M, Scott ME. Maternal gastrointestinal nematode infection enhances spatial memory of uninfected juvenile mouse pups. Sci Rep 2022; 12:9796. [PMID: 35697723 PMCID: PMC9192650 DOI: 10.1038/s41598-022-13971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022] Open
Abstract
The developing brain is particularly vulnerable to factors including maternal infection during pregnancy. Establishment of neural networks critical for memory and cognition begins during the perinatal period, when Heligmosomoides bakeri, a gastrointestinal (GI) nematode restricted to the maternal mouse intestine, has been shown to upregulate expression of long-term potentiation genes in the young rodent pup brain. We explored the impact of maternal infection during pregnancy and early lactation on the spatial behavior of uninfected male and female juvenile mice. Pre-weaned pups of H. bakeri infected dams exhibited less exploratory behaviour compared to pups of uninfected dams on postnatal day (PD) 16 but not PD 17, possibly reflecting a transient fear of an unfamiliar environment and/or a brief neurodevelopmental delay. Our two spatial memory tests show for the first time an enhancement of spatial memory in response to maternal nematode infection regardless of pup sex. At PD 17, pups of infected dams expressed object location memories after 3 h in the Object Location Test whereas offspring of uninfected mothers did not. In addition, at PD 34, juveniles of infected mothers retained their ability to find the escape hole in the Barnes Maze Test for one week whereas offspring from uninfected mothers did not. This finding is even more striking given that spatial memory was positively associated with pup length, yet this maternal infection impaired linear growth of pups. Thus, the positive impact of maternal infection on spatial memory countered any impairment associated with the shorter length of the pups. Overall, these novel findings indicate that a maternal GI nematode infection during pregnancy and lactation positively influences the spatial memory of uninfected juvenile offspring with potential fitness implications for the next generation.
Collapse
Affiliation(s)
- Sophia C Noel
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, Quebec, H9X 3V9, Canada
| | - Liana Fortin-Hamel
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, Quebec, H9X 3V9, Canada
| | - Manjurul Haque
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, Quebec, H9X 3V9, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, Quebec, H9X 3V9, Canada.
| |
Collapse
|
30
|
Rowe RK, Griesbach GS. Immune-endocrine interactions in the pathophysiology of sleep-wake disturbances following traumatic brain injury: A narrative review. Brain Res Bull 2022; 185:117-128. [DOI: 10.1016/j.brainresbull.2022.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 12/16/2022]
|
31
|
Honig MG, Del Mar NA, Henderson DL, O'Neal D, Yammanur M, Cox R, Li C, Perry AM, Moore BM, Reiner A. Raloxifene, a cannabinoid type-2 receptor inverse agonist, mitigates visual deficits and pathology and modulates microglia after ocular blast. Exp Eye Res 2022; 218:108966. [PMID: 35143834 DOI: 10.1016/j.exer.2022.108966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/04/2022] [Accepted: 01/24/2022] [Indexed: 11/19/2022]
Abstract
Visual deficits after ocular blast injury (OBI) are common, but pharmacological approaches to improve long-term outcomes have not been identified. Blast forces frequently damage the retina and optic nerves, and work on experimental animals has shown the pro-inflammatory actions of microglia can further exacerbate such injuries. Cannabinoid type-2 receptor (CB2) inverse agonists specifically target activated microglia, biasing them away from the harmful pro-inflammatory M1 state toward the helpful reparative M2 state. We previously found that treating mice with CB2 inverse agonists after traumatic brain injury, produced by either focal cranial air blast or dorsal cranial impact, greatly attenuated the visual deficits and pathology that otherwise resulted. Here we examined the consequences of single and repeat OBI and the benefit provided by raloxifene, an FDA-approved estrogen receptor drug that possesses noteworthy CB2 inverse agonism. After single OBI, although the amplitudes of the A- and B-waves of the electroretinogram and pupil light response appeared to be normal, the mice showed hints of deficits in contrast sensitivity and visual acuity, a trend toward optic nerve axon loss, and significantly increased light aversion, which were reversed by 2 weeks of daily treatment with raloxifene. Mice subjected to repeat OBI (5 blasts spaced 1 min apart), exhibited more severe visual deficits, including decreases in contrast sensitivity, visual acuity, the amplitudes of the A- and B-waves of the electroretinogram, light aversion, and resting pupil diameter (i.e. hyperconstriction), accompanied by the loss of photoreceptor cells and optic nerve axons, nearly all of which were mitigated by raloxifene. Interestingly, optic nerve axon abundance was strongly correlated with contrast sensitivity and visual acuity across all groups of experimental mice in the repeat OBI study, suggesting optic nerve axon loss with rOBI and its attenuation with raloxifene are associated with the extent of these two deficits while photoreceptor abundance was highly correlated with A-wave amplitude and resting pupil size, suggesting a prominent role for photoreceptors in these two deficits. Quantitative PCR (qPCR) showed levels of M1-type microglial markers (e.g. iNOS, IL1β, TNFα, and CD32) in retina, optic nerve, and thalamus were increased 3 days after repeat OBI. With raloxifene treatment, the overall expression of M1 markers was more similar to that in sham mice. Raloxifene treatment was also associated with the elevation of IL10 transcripts in all three tissues compared to repeat OBI alone, but the results for the three other M2 microglial markers we examined were more varied. Taken together, the qPCR results suggest that raloxifene benefit for visual function and pathology was associated with a lessening of the pro-inflammatory actions of microglia. The benefit we find for raloxifene following OBI provides a strong basis for phase-2 efficacy testing in human clinical trials for treating ocular injury.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Desmond L Henderson
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Dylan O'Neal
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Meghna Yammanur
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Rachel Cox
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Chunyan Li
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Aaron M Perry
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Bob M Moore
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology(,) the University of Tennessee Health Science Center, Memphis, TN, 38163, USA; Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
32
|
Perez-Rando M, Guirado R, Tellez-Merlo G, Carceller H, Nacher J. Estradiol Regulates Polysialylated Form of the Neural Cell Adhesion Molecule Expression and Connectivity of O-LM Interneurons in the Hippocampus of Adult Female Mice. Neuroendocrinology 2022; 112:51-67. [PMID: 33550289 DOI: 10.1159/000515052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/04/2021] [Indexed: 11/19/2022]
Abstract
The estrous cycle is caused by the changing concentration of ovarian hormones, particularly 17β-estradiol, a hormone whose effect on excitatory circuits has been extensively reported. However, fewer studies have tried to elucidate how this cycle, or this hormone, affects the plasticity of inhibitory networks and the structure of interneurons. Among these cells, somatostatin-expressing O-LM neurons of the hippocampus are especially interesting. They have a role in the modulation of theta oscillations, and they receive direct input from the entorhinal cortex, which place them in the center of hippocampal function. In this study, we report that the expression of polysialylated form of the neural cell adhesion molecule (PSA-NCAM) in the hippocampus, a molecule involved in the plasticity of somatostatin-expressing interneurons in the adult brain, fluctuated through the different stages of the estrous cycle. Likewise, these stages and the expression of PSA-NCAM affected the density of dendritic spines of O-LM cells. We also describe that 17β-estradiol replacement of adult ovariectomized female mice caused an increase in the perisomatic inhibitory puncta in O-LM interneurons as well as an increase in their axonal bouton density. Interestingly, this treatment also induced a decrease in their dendritic spine density, specifically in O-LM interneurons lacking PSA-NCAM expression. Finally, using an ex vivo real-time assay with entorhinal-hippocampal organotypic cultures, we show that this hormone decreased the dynamics in spinogenesis, altogether highlighting the modulatory effect that 17β-estradiol has on inhibitory circuits.
Collapse
Affiliation(s)
- Marta Perez-Rando
- Neurobiology Unit, Program in Neurosciences and BIOTECMED Institute, Universitat de València, Burjassot, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Ramon Guirado
- Neurobiology Unit, Program in Neurosciences and BIOTECMED Institute, Universitat de València, Burjassot, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
- Dirección General de Universidades, Gobierno de Aragón, Zaragoza, Spain
| | - Guillermina Tellez-Merlo
- Lab. Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Hector Carceller
- Neurobiology Unit, Program in Neurosciences and BIOTECMED Institute, Universitat de València, Burjassot, Spain
| | - Juan Nacher
- Neurobiology Unit, Program in Neurosciences and BIOTECMED Institute, Universitat de València, Burjassot, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
- CIBERSAM: Spanish National Network for Research in Mental Health, Valencia, Spain
| |
Collapse
|
33
|
Kuwahara N, Nicholson K, Isaacs L, MacLusky NJ. Androgen Effects on Neural Plasticity. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:216-230. [PMID: 35024693 PMCID: PMC8744448 DOI: 10.1089/andro.2021.0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Androgens are synthesized in the brain, gonads, and adrenal glands, in both sexes, exerting physiologically important effects on the structure and function of the central nervous system. These effects may contribute to the incidence and progression of neurological disorders such as autism spectrum disorder, schizophrenia, and Alzheimer's disease, which occur at different rates in males and females. This review briefly summarizes the current state of knowledge with respect to the neuroplastic effects of androgens, with particular emphasis on the hippocampus, which has been the focus of much of the research in this field.
Collapse
Affiliation(s)
- Nariko Kuwahara
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kate Nicholson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Lauren Isaacs
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Neil J. MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
34
|
Ceccarelli I, Bioletti L, Peparini S, Solomita E, Ricci C, Casini I, Miceli E, Aloisi AM. Estrogens and phytoestrogens in body functions. Neurosci Biobehav Rev 2021; 132:648-663. [PMID: 34890602 DOI: 10.1016/j.neubiorev.2021.12.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/23/2022]
Abstract
Estrogens are the hormones of reproduction in women as well as of many other important functions in the male and female body. They undergo significant changes in the different phases of life, e.g. during puberty, pregnancy or at menopause/andropause. Phytoestrogens are natural non-steroidal phenolic plant compounds that can mimic the activity of estrogens and their beneficial effects in women and in men. This narrative review summarizes the literature on the physiological role of estrogens and the several potential health benefits of phytoestrogens, with particular attention given to the possible role of phytoestrogens in aging.
Collapse
Affiliation(s)
- Ilaria Ceccarelli
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Lucia Bioletti
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Sofia Peparini
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Erminia Solomita
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Comasia Ricci
- Department Life Sciences, University of Siena, Siena, Italy
| | - Ilenia Casini
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Elisangela Miceli
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Anna Maria Aloisi
- Department Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.
| |
Collapse
|
35
|
Honig MG, Del Mar NA, Henderson DL, O'Neal D, Doty JB, Cox R, Li C, Perry AM, Moore BM, Reiner A. Raloxifene Modulates Microglia and Rescues Visual Deficits and Pathology After Impact Traumatic Brain Injury. Front Neurosci 2021; 15:701317. [PMID: 34776838 PMCID: PMC8585747 DOI: 10.3389/fnins.2021.701317] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/07/2021] [Indexed: 11/29/2022] Open
Abstract
Mild traumatic brain injury (TBI) involves widespread axonal injury and activation of microglia, which initiates secondary processes that worsen the TBI outcome. The upregulation of cannabinoid type-2 receptors (CB2) when microglia become activated allows CB2-binding drugs to selectively target microglia. CB2 inverse agonists modulate activated microglia by shifting them away from the harmful pro-inflammatory M1 state toward the helpful reparative M2 state and thus can stem secondary injury cascades. We previously found that treatment with the CB2 inverse agonist SMM-189 after mild TBI in mice produced by focal cranial blast rescues visual deficits and the optic nerve axon loss that would otherwise result. We have further shown that raloxifene, which is Food and Drug Administration (FDA)-approved as an estrogen receptor modulator to treat osteoporosis, but also possesses CB2 inverse agonism, yields similar benefit in this TBI model through its modulation of microglia. As many different traumatic events produce TBI in humans, it is widely acknowledged that diverse animal models must be used in evaluating possible therapies. Here we examine the consequences of TBI created by blunt impact to the mouse head for visual function and associated pathologies and assess raloxifene benefit. We found that mice subjected to impact TBI exhibited decreases in contrast sensitivity and the B-wave of the electroretinogram, increases in light aversion and resting pupil diameter, and optic nerve axon loss, which were rescued by daily injection of raloxifene at 5 or 10 mg/ml for 2 weeks. Raloxifene treatment was associated with reduced M1 activation and/or enhanced M2 activation in retina, optic nerve, and optic tract after impact TBI. Our results suggest that the higher raloxifene dose, in particular, may be therapeutic for the optic nerve by enhancing the phagocytosis of axonal debris that would otherwise promote inflammation, thereby salvaging less damaged axons. Our current work, together with our prior studies, shows that microglial activation drives secondary injury processes after both impact and cranial blast TBI and raloxifene mitigates microglial activation and visual system injury in both cases. The results thus provide a strong basis for phase 2 human clinical trials evaluating raloxifene as a TBI therapy.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Desmond L Henderson
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Dylan O'Neal
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - John B Doty
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Rachel Cox
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chunyan Li
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Aaron M Perry
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Bob M Moore
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
36
|
Soutar CN, Grenier P, Patel A, Kabitsis PP, Olmstead MC, Bailey CDC, Dringenberg HC. Brain-Generated 17β-Estradiol Modulates Long-Term Synaptic Plasticity in the Primary Auditory Cortex of Adult Male Rats. Cereb Cortex 2021; 32:2140-2155. [PMID: 34628498 DOI: 10.1093/cercor/bhab345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Neuron-derived 17β-estradiol (E2) alters synaptic transmission and plasticity in brain regions with endocrine and non-endocrine functions. Investigations into a modulatory role of E2 in synaptic activity and plasticity have mainly focused on the rodent hippocampal formation. In songbirds, E2 is synthesized by auditory forebrain neurons and promotes auditory signal processing and memory for salient acoustic stimuli; however, the modulatory effects of E2 on memory-related synaptic plasticity mechanisms have not been directly examined in the auditory forebrain. We investigated the effects of bidirectional E2 manipulations on synaptic transmission and long-term potentiation (LTP) in the rat primary auditory cortex (A1). Immunohistochemistry revealed widespread neuronal expression of the E2 biosynthetic enzyme aromatase in multiple regions of the rat sensory and association neocortex, including A1. In A1, E2 application reduced the threshold for in vivo LTP induction at layer IV synapses, whereas pharmacological suppression of E2 production by aromatase inhibition abolished LTP induction at layer II/III synapses. In acute A1 slices, glutamate and γ-aminobutyric acid (GABA) receptor-mediated currents were sensitive to E2 manipulations in a layer-specific manner. These findings demonstrate that locally synthesized E2 modulates synaptic transmission and plasticity in A1 and suggest potential mechanisms by which E2 contributes to auditory signal processing and memory.
Collapse
Affiliation(s)
- Chloe N Soutar
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Patrick Grenier
- Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Ashutosh Patel
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Pauline P Kabitsis
- Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Mary C Olmstead
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada.,Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Craig D C Bailey
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Hans C Dringenberg
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada.,Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| |
Collapse
|
37
|
Savaşçı BB, Lucon-Xiccato T, Bisazza A. Ontogeny and personality affect inhibitory control in guppies, Poecilia reticulata. Anim Behav 2021. [DOI: 10.1016/j.anbehav.2021.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
38
|
López-Olmeda JF, Zhao H, Reischl M, Pylatiuk C, Lucon-Xiccato T, Loosli F, Foulkes NS. Long photoperiod impairs learning in male but not female medaka. iScience 2021; 24:102784. [PMID: 34308290 PMCID: PMC8283132 DOI: 10.1016/j.isci.2021.102784] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/12/2021] [Accepted: 06/23/2021] [Indexed: 01/01/2023] Open
Abstract
Day length in conjunction with seasonal cycles affects many aspects of animal biology. We have studied photoperiod-dependent alterations of complex behavior in the teleost, medaka (Oryzias latipes), a photoperiodic breeder, in a learning paradigm whereby fish have to activate a sensor to obtain a food reward. Medaka were tested under a long (14:10 LD) and short (10:14 LD) photoperiod in three different groups: mixed-sex, all-males, and all-females. Under long photoperiod, medaka mixed-sex groups learned rapidly with a stable response. Unexpectedly, males-only groups showed a strong learning deficit, whereas females-only groups performed efficiently. In mixed-sex groups, female individuals drove group learning, whereas males apparently prioritized mating over feeding behavior resulting in strongly reduced learning performance. Under short photoperiod, where medaka do not mate, male performance improved to a level similar to that of females. Thus, photoperiod has sex-specific effects on the learning performance of a seasonal vertebrate.
Collapse
Affiliation(s)
- Jose Fernando López-Olmeda
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
- Department of Physiology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain
| | - Haiyu Zhao
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
- School of Life Sciences, Lanzhou University, No.222 South Tianshui Road, 730000 Lanzhou, PR China
| | - Markus Reischl
- Institute for Applied Computer Science, Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Christian Pylatiuk
- Institute for Applied Computer Science, Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Tyrone Lucon-Xiccato
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Felix Loosli
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Nicholas S. Foulkes
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
39
|
Intraperitoneal Carbamylated erythropoietin improves memory and hippocampal apoptosis in beta amyloid rat model of Alzheimer’s disease through stimulating autophagy and inhibiting necroptosis. PHYSIOLOGY AND PHARMACOLOGY 2021. [DOI: 10.52547/phypha.26.4.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
40
|
Bouton ME, Maren S, McNally GP. BEHAVIORAL AND NEUROBIOLOGICAL MECHANISMS OF PAVLOVIAN AND INSTRUMENTAL EXTINCTION LEARNING. Physiol Rev 2021; 101:611-681. [PMID: 32970967 PMCID: PMC8428921 DOI: 10.1152/physrev.00016.2020] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This article reviews the behavioral neuroscience of extinction, the phenomenon in which a behavior that has been acquired through Pavlovian or instrumental (operant) learning decreases in strength when the outcome that reinforced it is removed. Behavioral research indicates that neither Pavlovian nor operant extinction depends substantially on erasure of the original learning but instead depends on new inhibitory learning that is primarily expressed in the context in which it is learned, as exemplified by the renewal effect. Although the nature of the inhibition may differ in Pavlovian and operant extinction, in either case the decline in responding may depend on both generalization decrement and the correction of prediction error. At the neural level, Pavlovian extinction requires a tripartite neural circuit involving the amygdala, prefrontal cortex, and hippocampus. Synaptic plasticity in the amygdala is essential for extinction learning, and prefrontal cortical inhibition of amygdala neurons encoding fear memories is involved in extinction retrieval. Hippocampal-prefrontal circuits mediate fear relapse phenomena, including renewal. Instrumental extinction involves distinct ensembles in corticostriatal, striatopallidal, and striatohypothalamic circuits as well as their thalamic returns for inhibitory (extinction) and excitatory (renewal and other relapse phenomena) control over operant responding. The field has made significant progress in recent decades, although a fully integrated biobehavioral understanding still awaits.
Collapse
Affiliation(s)
- Mark E Bouton
- Department of Psychological Science, University of Vermont, Burlington, Vermont
| | - Stephen Maren
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, Texas
| | - Gavan P McNally
- School of Psychology, University of New South Wales, Sydney, Australia
| |
Collapse
|
41
|
Alonso A, Genzel L, Gomez A. Sex and Menstrual Phase Influences on Sleep and Memory. CURRENT SLEEP MEDICINE REPORTS 2021. [DOI: 10.1007/s40675-020-00201-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Abstract
Purposes of Review
This review highlights the effect of sex differences in sleep mediated memory consolidation and cognitive performance. In addition, the role of menstrual cycle and the fluctuating level of sexual hormones (mainly oestrogen and progesterone) are stressed.
Recent Findings
The literature indicates that sex hormones mediate and orchestrate the differences observed in performance of females in comparison with males in a variety of tasks and can also be related to how sleep benefits cognition. Although the exact mechanism of such influence is not clear, it most likely involves differential activation of brain areas, sensitivity to neuromodulators (mainly oestrogen), circadian regulation of sleep and temperature, as well as modification of strategies to solve tasks across the menstrual cycle.
Summary
With the evidence presented here, we hope to encourage researchers to develop appropriate paradigms to study the complex relationship between menstrual cycle, sleep (its regulation, architecture and electrophysiological hallmarks) and performance in memory and other cognitive tasks.
Collapse
|
42
|
Intact Female Mice Acquire Trace Eyeblink Conditioning Faster than Male and Ovariectomized Female Mice. eNeuro 2021; 8:ENEURO.0199-20.2021. [PMID: 33531367 PMCID: PMC7986530 DOI: 10.1523/eneuro.0199-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 12/04/2020] [Accepted: 01/06/2021] [Indexed: 11/21/2022] Open
Abstract
Female subjects have been widely excluded from past neuroscience work because of a number of biases, including the notion that cycling sex hormones increase variability. However, it is necessary to conduct behavioral research in mice that includes both sexes as mice are typically used for developing and evaluating future therapeutics. Understanding sex differences in learning is fundamental for the development of targeted therapies for numerous neurologic and neurodegenerative disorders, including Alzheimer’s disease, which is more prevalent in females than males. This study set out to confirm the role of sex and necessity of circulating ovarian hormones in the acquisition of the temporal associative memory task trace eyeblink conditioning (tEBC) in C57BL/6J mice. We present evidence that sex and ovarian hormones are important factors in learning. Specifically, intact female mice learn significantly faster than both male and ovariectomized (ovx) female mice. Data from pseudoconditioned control mice indicate that sex differences are because of differences in learned associations, not sensitization or spontaneous blink rate. This study strengthens the idea that ovarian hormones such as estrogen and progesterone significantly influence learning and memory and that further research is needed to determine the underlying mechanisms behind their effects. Overall, our findings emphasize the necessity of including both sexes in future behavioral studies.
Collapse
|
43
|
Wang J, Li HY, Shen SY, Zhang JR, Liang LF, Huang HJ, Li B, Wu GC, Zhang YQ, Yu J. The antidepressant and anxiolytic effect of GPER on translocator protein (TSPO) via protein kinase a (PKA) signaling in menopausal female rats. J Steroid Biochem Mol Biol 2021; 207:105807. [PMID: 33345973 DOI: 10.1016/j.jsbmb.2020.105807] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 11/05/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022]
Abstract
Postmenopausal depression is mainly caused by the deprivation of ovarian hormones during menopausal transition, it is of great importance to study on the treatment that could effectively relieve symptoms of menopausal depression with fewer side effects. Activation of G-protein-coupled estrogen receptor (GPER) has long been reported to facilitate neuronal plasticity and improve cognition in animals. Meanwhile, it could participate in regulation of intracellular signaling pathways through the characteristic of GPER, ameliorate intracellular mitochondrial function and oxidative stress. However, the impact of GPER on regulating estrogen deprived-depressant and anxious behaviors is still largely unknown. Here we used the ovariectomized female rats to imitate the condition of menopause. Owing to the lateral ventricle administration of G-1 which specifically react with GPER receptor intracerebrally, Ovariectomized (OVX) female rats showed depressive- or anxiety-like phenotypes with attenuated mitochondrial function. In addition, G-1 facilitated PKA activation, which further accelerated TSPO phosphorylation and alleviated menopausal depression- and anxiety-like behaviors. Moreover, PKA inhibitor PKI could partially antagonized the anti-anxiety and anti-depression effects of G-1. Taken together, we concluded that GPER activation might exhibit antidepressant and anxiolytic effect by elevating TSPO phosphorylation via protein kinase A signaling and rescuing the redox status in menopausal female rats.
Collapse
Affiliation(s)
- Jing Wang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hao-Yuan Li
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shi-Yu Shen
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jia-Rui Zhang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ling-Feng Liang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hui-Jie Huang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Bing Li
- Center Laboratories, Jinshan Hospital of Fudan University, Shanghai, 201508, China
| | - Gen-Cheng Wu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yu-Qiu Zhang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
44
|
Devulapalli R, Jones N, Farrell K, Musaus M, Kugler H, McFadden T, Orsi SA, Martin K, Nelsen J, Navabpour S, O'Donnell M, McCoig E, Jarome TJ. Males and females differ in the regulation and engagement of, but not requirement for, protein degradation in the amygdala during fear memory formation. Neurobiol Learn Mem 2021; 180:107404. [PMID: 33609735 DOI: 10.1016/j.nlm.2021.107404] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/28/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Over the last decade, strong evidence has emerged that protein degradation mediated by the ubiquitin-proteasome system is critical for fear memory formation in the amygdala. However, this work has been done primarily in males, leaving unanswered questions about whether females also require protein degradation during fear memory formation. Here, we found that male and female rats differed in their engagement and regulation of, but not need for, protein degradation in the amygdala during fear memory formation. Male, but not female, rats had increased protein degradation in the nuclei of amygdala cells after fear conditioning. Conversely, females had elevated baseline levels of overall ubiquitin-proteasome activity in amygdala nuclei. Gene expression and DNA methylation analyses identified that females had increased baseline expression of the ubiquitin coding gene Uba52, which had increased DNA 5-hydroxymethylation (5hmc) in its promoter region, indicating a euchromatin state necessary for increased levels of ubiquitin in females. Consistent with this, persistent CRISPR-dCas9 mediated silencing of Uba52 and proteasome subunit Psmd14 in the amygdala reduced baseline protein degradation levels and impaired fear memory in male and female rats, while enhancing baseline protein degradation in the amygdala of both sexes promoted fear memory formation. These results suggest that while both males and females require protein degradation in the amygdala for fear memory formation, they differ in their baseline regulation and engagement of this process following learning. These results have important implications for understanding the etiology of sex-related differences in fear memory formation.
Collapse
Affiliation(s)
- Rishi Devulapalli
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Natalie Jones
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Kayla Farrell
- Department of Animal and Poultry Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Madeline Musaus
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Hannah Kugler
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Taylor McFadden
- Department of Animal and Poultry Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Sabrina A Orsi
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Kiley Martin
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Jacob Nelsen
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Shaghayegh Navabpour
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | - Madison O'Donnell
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Emmarose McCoig
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Timothy J Jarome
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; Department of Animal and Poultry Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, USA.
| |
Collapse
|
45
|
Holden MP, Hampson E. Endogenous variation in estradiol in women affects the weighting of metric and categorical information in spatial location memory. Horm Behav 2021; 128:104909. [PMID: 33279507 DOI: 10.1016/j.yhbeh.2020.104909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022]
Abstract
Recent work has suggested that sex differences may exist in the strategies or types of cues that are utilized by men and women to remember discrete spatial locations or routes through a visual environment. The current study investigated the effects of circulating estradiol levels in women on the relative weighting of categorical versus fine-grained 'metric' information in a test of short-term memory for spatial locations, either presented within a simple geometric surround (a circular enclosure) or within more visually complex landscape scenes. Patterns of displacement error in the point location estimates made by men and women were analyzed. Results confirmed a sex difference in the weighting of metric versus categorical cues. Relative to men, women's estimates of locations were more strongly biased toward the center of the surrounding category (i.e., toward the category 'prototype'). Furthermore, objective measures of estradiol via saliva collected at the time of memory testing showed that, among naturally-cycling women, estradiol concentrations correlated in a positive, graded, fashion with the degree of emphasis that women placed on categorical information when estimating point locations. No associations were found for progesterone. These findings are consistent with a wider body of research showing that biological sex and reproductive hormone levels, including 17β-estradiol, can subtly influence performance on certain spatial tasks. This is the first study to show that circulating estradiol levels may influence the relative emphasis placed on categorical versus metric cues when remembering simple point locations.
Collapse
Affiliation(s)
- Mark P Holden
- Department of Psychology, University of Western Ontario, London, Ontario N6G 3V1, Canada.
| | - Elizabeth Hampson
- Department of Psychology, University of Western Ontario, London, Ontario N6G 3V1, Canada
| |
Collapse
|
46
|
Hokenson RE, Short AK, Chen Y, Pham AL, Adams ET, Bolton JL, Swarup V, Gall CM, Baram TZ. Unexpected Role of Physiological Estrogen in Acute Stress-Induced Memory Deficits. J Neurosci 2021; 41:648-662. [PMID: 33262247 PMCID: PMC7842761 DOI: 10.1523/jneurosci.2146-20.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 11/22/2022] Open
Abstract
Stress may promote emotional and cognitive disturbances, which differ by sex. Adverse outcomes, including memory disturbances, are typically observed following chronic stress, but are now being recognized also after short events, including mass shootings, assault, or natural disasters, events that consist of concurrent multiple acute stresses (MAS). Prior work has established profound and enduring effects of MAS on memory in males. Here we examined the effects of MAS on female mice and probed the role of hormonal fluctuations during the estrous cycle on MAS-induced memory problems and the underlying brain network and cellular mechanisms. Female mice were impacted by MAS in an estrous cycle-dependent manner: MAS impaired hippocampus-dependent spatial memory in early-proestrous mice, characterized by high levels of estradiol, whereas memory of mice stressed during estrus (low estradiol) was spared. As spatial memory requires an intact dorsal hippocampal CA1, we examined synaptic integrity in mice stressed at different cycle phases and found a congruence of dendritic spine density and spatial memory deficits, with reduced spine density only in mice stressed during high estradiol cycle phases. Assessing MAS-induced activation of brain networks interconnected with hippocampus, we identified differential estrous cycle-dependent activation of memory- and stress-related regions, including the amygdala. Network analyses of the cross-correlation of fos expression among these regions uncovered functional connectivity that differentiated impaired mice from those not impaired by MAS. In conclusion, the estrous cycle modulates the impact of MAS on spatial memory, and fluctuating physiological levels of sex hormones may contribute to this effect.SIGNIFICANCE STATEMENT: Effects of stress on brain functions, including memory, are profound and sex-dependent. Acute stressors occurring simultaneously result in spatial memory impairments in males, but effects on females are unknown. Here we identified estrous cycle-dependent effects of such stresses on memory in females. Surprisingly, females with higher physiological estradiol experienced stress-induced memory impairment and a loss of underlying synapses. Memory- and stress-responsive brain regions interconnected with hippocampus were differentially activated across high and low estradiol mice, and predicted memory impairment. Thus, at functional, network, and cellular levels, physiological estradiol influences the effects of stress on memory in females, providing insight into mechanisms of prominent sex differences in stress-related memory disorders, such as post-traumatic stress disorder.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christine M Gall
- Departments of Anatomy and Neurobiology
- Neurobiology and Behavior
| | - Tallie Z Baram
- Departments of Anatomy and Neurobiology
- Pediatrics
- Neurology, University of California-Irvine, Irvine, California 92697
| |
Collapse
|
47
|
Taylor CM, Pritschet L, Jacobs EG. The scientific body of knowledge - Whose body does it serve? A spotlight on oral contraceptives and women's health factors in neuroimaging. Front Neuroendocrinol 2021; 60:100874. [PMID: 33002517 PMCID: PMC7882021 DOI: 10.1016/j.yfrne.2020.100874] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
Women constitute half of the world's population, yet neuroscience research does not serve the sexes equally. Fifty years of preclinical animal evidence documents the tightly-coupled relationship between our endocrine and nervous systems, yet human neuroimaging studies rarely consider how endocrine factors shape the structural and functional architecture of the human brain. Here, we quantify several blind spots in neuroimaging research, which overlooks aspects of the human condition that impact women's health (e.g. the menstrual cycle, hormonal contraceptives, pregnancy, menopause). Next, we illuminate potential consequences of this oversight: today over 100 million women use oral hormonal contraceptives, yet relatively few investigations have systematically examined whether disrupting endogenous hormone production impacts the brain. We close by presenting a roadmap for progress, highlighting the University of California Women's Brain Initiative which is addressing unmet needs in women's health research.
Collapse
Affiliation(s)
- Caitlin M Taylor
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, United States.
| | - Laura Pritschet
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, United States
| | - Emily G Jacobs
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, United States; Neuroscience Research Institute, University of California, Santa Barbara, United States.
| |
Collapse
|
48
|
Hill RA, Kouremenos K, Tull D, Maggi A, Schroeder A, Gibbons A, Kulkarni J, Sundram S, Du X. Bazedoxifene - a promising brain active SERM that crosses the blood brain barrier and enhances spatial memory. Psychoneuroendocrinology 2020; 121:104830. [PMID: 32858306 DOI: 10.1016/j.psyneuen.2020.104830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022]
Abstract
Over 20 years of accumulated evidence has shown that the major female sex hormone 17β-estradiol can enhance cognitive functioning. However, the utility of estradiol as a therapeutic cognitive enhancer is hindered by its unwanted peripheral effects (carcinogenic). Selective estrogen receptor modulators (SERMs) avoid the unwanted effects of estradiol by acting as estrogen receptor antagonists in some tissues such as breast and uterus, but as agonists in others such as bone, and are currently used for the treatment of osteoporosis. However, understanding of their actions in the brain are limited. The third generation SERM bazedoxifene has recently been FDA approved for clinical use with an improved biosafety profile. However, whether bazedoxifene can enter the brain and enhance cognition is unknown. Using mice, the current study aimed to explore if bazedoxifene can 1) cross the blood-brain barrier, 2) rescue ovariectomy-induced hippocampal-dependent spatial memory deficit, and 3) activate neural estrogen response element (ERE)-dependent gene transcription. Using liquid chromatography-mass spectrometry (LC-MS), we firstly demonstrate that a peripheral injection of bazedoxifene can enter the brain. Secondly, we show that an acute intraperitoneal injection of bazedoxifene can rescue ovariectomy-induced spatial memory deficits. And finally, using the ERE-luciferase reporter mouse, we show in vivo that bazedoxifene can activate the ERE in the brain. The evidence shown here suggest bazedoxifene could be a viable cognitive enhancer with promising clinical applicability.
Collapse
Affiliation(s)
- R A Hill
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia; Florey Institute for Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.
| | - K Kouremenos
- Metabolomics Australia, Bio21 Molecular Science & Biotechnology Institute, Parkville, VIC, 3052, Australia
| | - D Tull
- Metabolomics Australia, Bio21 Molecular Science & Biotechnology Institute, Parkville, VIC, 3052, Australia
| | - A Maggi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, 20133, Italy
| | - A Schroeder
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia
| | - A Gibbons
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia
| | - J Kulkarni
- Monash Alfred Psychiatry Research Centre, Monash University, St Kilda, VIC, 3004, Australia
| | - S Sundram
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia
| | - X Du
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia; Florey Institute for Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| |
Collapse
|
49
|
Taxier LR, Gross KS, Frick KM. Oestradiol as a neuromodulator of learning and memory. Nat Rev Neurosci 2020; 21:535-550. [PMID: 32879508 PMCID: PMC8302223 DOI: 10.1038/s41583-020-0362-7] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2020] [Indexed: 12/24/2022]
Abstract
Although hormones such as glucocorticoids have been broadly accepted in recent decades as general neuromodulators of memory processes, sex steroid hormones such as the potent oestrogen 17β-oestradiol have been less well recognized by the scientific community in this capacity. The predominance of females in studies of oestradiol and memory and the general (but erroneous) perception that oestrogens are 'female' hormones have probably prevented oestradiol from being more widely considered as a key memory modulator in both sexes. Indeed, although considerable evidence supports a crucial role for oestradiol in regulating learning and memory in females, a growing body of literature indicates a similar role in males. This Review discusses the mechanisms of oestradiol signalling and provides an overview of the effects of oestradiol on spatial, object recognition, social and fear memories. Although the primary focus is on data collected in females, effects of oestradiol on memory in males will be discussed, as will sex differences in the molecular mechanisms that regulate oestrogenic modulation of memory, which may have important implications for the development of future cognitive therapeutics.
Collapse
Affiliation(s)
- Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| |
Collapse
|
50
|
Schwabe MR, Taxier LR, Frick KM. It takes a neural village: Circuit-based approaches for estrogenic regulation of episodic memory. Front Neuroendocrinol 2020; 59:100860. [PMID: 32781195 PMCID: PMC7669700 DOI: 10.1016/j.yfrne.2020.100860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
Cognitive behaviors, such as episodic memory formation, are complex processes involving coordinated activity in multiple brain regions. However, much of the research on hormonal regulation of cognition focuses on manipulation of one region at a time or provides a single snapshot of how a systemic treatment affects multiple brain regions without investigating how these regions might interact to mediate hormone effects. Here, we use estrogenic regulation of episodic memory as an example of how circuit-based approaches may be incorporated into future studies of hormones and cognition. We first review basic episodic memory circuitry, rapid mechanisms by which 17β-estradiol can alter circuit activity, and current knowledge about 17β-estradiol's effects on episodic memory. Next, we outline approaches that researchers can employ to consider circuit effects in their estrogen research and provide examples of how these methods have been used to examine hormonal regulation of memory and other behaviors.
Collapse
Affiliation(s)
- Miranda R Schwabe
- 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
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|