1
|
Bencker C, Gschwandtner L, Nayman S, Grikšienė R, Nguyen B, Nater UM, Guennoun R, Sundström-Poromaa I, Pletzer B, Bixo M, Comasco E. Progestagens and progesterone receptor modulation: Effects on the brain, mood, stress, and cognition in females. Front Neuroendocrinol 2024; 76:101160. [PMID: 39515587 DOI: 10.1016/j.yfrne.2024.101160] [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: 08/19/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Progesterone is a highly lipophilic gonadal hormone that can influence behavior and mental health through its receptors in the brain. Fluctuations in progesterone levels across critical periods of a females life are associated with increased susceptibility to mental conditions. This review highlights the effects of progestagens, including progesterone and synthetic progestins, on the brain, mood, stress, and cognition in females. The primary focus is on experimental pharmacological research that teases out the distinct effects of progestagens from those of estrogens. Additionally, the key literature on puberty, the menstrual cycle, pregnancy, perimenopause, hormonal contraceptives, and menopausal hormone therapy is reviewed, although conclusions are limited by the nested effects of progestagens and estrogens. Single study-findings suggest an influence of progesterone on amygdala reactivity related to processing of emotional stimuli and memory. In patients with premenstrual dysphoric disorder, progesterone receptor modulation improves premenstrual mood symptoms and potentially enhances fronto-cingulate control over emotion processing. The interaction between progestagens and the systems involved in the regulation of stress seems to influence subjective experiences of mood and stress. Sparse studies investigating the effects of progestin-only contraceptives suggest effects of progestagens on the brain, mood, and stress. Progesterone and progestins used for contraception can influence neural processes as myelination and neuroprotection, exerting protective effects against stroke. Concerning menopausal hormonal therapy, the effects of progestins are largely unknown. Levels of progesterone as well as type, administration route, timing, dose regimen, metabolism, and intracellular activity of progestins in hormonal contraceptives and menopausal hormonal therapy are factors whose effects remain to be elucidated. Altogether, current knowledge highlights the potential role of progestagens in females health but also calls for well-designed pharmaco-behavioral studies disentangling the effects of progestagens from those of estrogens.
Collapse
Affiliation(s)
- Celine Bencker
- Department of Clinical and Health Psychology, Faculty of Psychology, University of Vienna, Vienna, Austria; University Research Platform "Stress of Life (SOLE) - Processes and Mechanisms underlying Everyday Life Stress", University of Vienna, Vienna, Austria
| | - Laura Gschwandtner
- Department of Clinical and Health Psychology, Faculty of Psychology, University of Vienna, Vienna, Austria; University Research Platform "Stress of Life (SOLE) - Processes and Mechanisms underlying Everyday Life Stress", University of Vienna, Vienna, Austria
| | - Sibel Nayman
- Research Group Longitudinal and Intervention Research, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, Mannheim, Germany
| | - Ramunė Grikšienė
- Department of Neurobiology and Biophysics, Life Science Center, Vilnius University, Lithuania
| | | | - Urs M Nater
- Department of Clinical and Health Psychology, Faculty of Psychology, University of Vienna, Vienna, Austria; University Research Platform "Stress of Life (SOLE) - Processes and Mechanisms underlying Everyday Life Stress", University of Vienna, Vienna, Austria
| | | | | | - Belinda Pletzer
- Department of Psychology, Centre for Cognitive Neuroscience, University of Salzburg, Austria
| | - Marie Bixo
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Sweden
| | - Erika Comasco
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Sweden.
| |
Collapse
|
2
|
Chen MH, Lin HC, Chao T, Lee VSY, Hou CL, Wang TJ, Chen JR. Hyaluronic Acid Conjugated with 17β-Estradiol Effectively Alleviates Estropause-Induced Cognitive Deficits in Rats. Int J Mol Sci 2023; 24:15569. [PMID: 37958552 PMCID: PMC10649161 DOI: 10.3390/ijms242115569] [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: 09/28/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Women are at a higher risk of cognitive impairments and Alzheimer's disease (AD), particularly after the menopause, when the estrous cycle becomes irregular and diminishes. Numerous studies have shown that estrogen deficiency, especially estradiol (E2) deficiency, plays a key role in this phenomenon. Recently, a novel polymeric drug, hyaluronic acid-17β-estradiol conjugate (HA-E2), has been introduced for the delivery of E2 to brain tissues. Studies have indicated that HA-E2 crosses the blood-brain barrier (BBB) and facilitates a prolonged E2 release profile while lowering the risk of estrogen-supplement-related side effects. In this study, we used ovariohysterectomy (OHE) rats, a postmenopausal cognitive deficit model, to explore the effect of a 2-week HA-E2 treatment (210 ng/kg body weight, twice a week) on the cholinergic septo-hippocampal innervation system, synaptic transmission in hippocampal pyramidal neurons and cognitive improvements. Our study revealed an 11% rise in choline acetyltransferase (ChAT) expression in both the medial septal nucleus (MS nucleus) and the hippocampus, along with a 14-18% increase in dendritic spine density in hippocampal pyramidal neurons, following HA-E2 treatment in OHE rats. These enhancements prompted the recovery of cognitive functions such as spatial learning and memory. These findings suggest that HA-E2 may prevent and improve estrogen-deficiency-induced cognitive impairment and AD.
Collapse
Affiliation(s)
- Mu-Hsuan Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, No. 145, Xingda Rd., Taichung 402202, Taiwan; (M.-H.C.); (H.-C.L.); (T.C.)
| | - Hsiao-Chun Lin
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, No. 145, Xingda Rd., Taichung 402202, Taiwan; (M.-H.C.); (H.-C.L.); (T.C.)
| | - Tzu Chao
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, No. 145, Xingda Rd., Taichung 402202, Taiwan; (M.-H.C.); (H.-C.L.); (T.C.)
| | - Viola Szu-Yuan Lee
- Basic Research Division, Holy Stone Healthcare Co., Ltd., Taipei 11493, Taiwan; (V.S.-Y.L.); (C.-L.H.)
| | - Chia-Lung Hou
- Basic Research Division, Holy Stone Healthcare Co., Ltd., Taipei 11493, Taiwan; (V.S.-Y.L.); (C.-L.H.)
| | - Tsyr-Jiuan Wang
- Department of Nursing, National Taichung University of Science and Technology, No. 193, Section 1, Sanmin Rd., Taichung 403027, Taiwan
| | - Jeng-Rung Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, No. 145, Xingda Rd., Taichung 402202, Taiwan; (M.-H.C.); (H.-C.L.); (T.C.)
| |
Collapse
|
3
|
Daniel JM, Lindsey SH, Mostany R, Schrader LA, Zsombok A. Cardiometabolic health, menopausal estrogen therapy and the brain: How effects of estrogens diverge in healthy and unhealthy preclinical models of aging. Front Neuroendocrinol 2023; 70:101068. [PMID: 37061205 PMCID: PMC10725785 DOI: 10.1016/j.yfrne.2023.101068] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023]
Abstract
Research in preclinical models indicates that estrogens are neuroprotective and positively impact cognitive aging. However, clinical data are equivocal as to the benefits of menopausal estrogen therapy to the brain and cognition. Pre-existing cardiometabolic disease may modulate mechanisms by which estrogens act, potentially reducing or reversing protections they provide against cognitive decline. In the current review we propose mechanisms by which cardiometabolic disease may alter estrogen effects, including both alterations in actions directly on brain memory systems and actions on cardiometabolic systems, which in turn impact brain memory systems. Consideration of mechanisms by which estrogen administration can exert differential effects dependent upon health phenotype is consistent with the move towards precision or personalized medicine, which aims to determine which treatment interventions will work for which individuals. Understanding effects of estrogens in both healthy and unhealthy models of aging is critical to optimizing the translational link between preclinical and clinical research.
Collapse
Affiliation(s)
- Jill M Daniel
- Department of Psychology and Brain Institute, Tulane University, New Orleans, LA, United States.
| | - Sarah H Lindsey
- Department of Pharmacology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Ricardo Mostany
- Department of Pharmacology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Laura A Schrader
- Department of Cell & Molecular Biology and Brain Institute, Tulane University, New Orleans, LA, United States
| | - Andrea Zsombok
- Department of Physiology and Brain Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
4
|
Wilson KM, Arquilla AM, Saltzman W. The parental umwelt: Effects of parenthood on sensory processing in rodents. J Neuroendocrinol 2023; 35:e13237. [PMID: 36792373 DOI: 10.1111/jne.13237] [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: 10/31/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Abstract
An animal's umwelt, comprising its perception of the sensory environment, which is inherently subjective, can change across the lifespan in accordance with major life events. In mammals, the onset of motherhood, in particular, is associated with a neural and sensory plasticity that alters a mother's detection and use of sensory information such as infant-related sensory stimuli. Although the literature surrounding mammalian mothers is well established, very few studies have addressed the effects of parenthood on sensory plasticity in mammalian fathers. In this review, we summarize the major findings on the effects of parenthood on behavioural and neural responses to sensory stimuli from pups in rodent mothers, with a focus on the olfactory, auditory, and somatosensory systems, as well as multisensory integration. We also review the available literature on sensory plasticity in rodent fathers. Finally, we discuss the importance of sensory plasticity for effective parental care, hormonal modulation of plasticity, and an exploration of temporal, ecological, and life-history considerations of sensory plasticity associated with parenthood. The changes in processing and/or perception of sensory stimuli associated with the onset of parental care may have both transient and long-lasting effects on parental behaviour and cognition in both mothers and fathers; as such, several promising areas of study, such as on the molecular/genetic, neurochemical, and experiential underpinnings of parenthood-related sensory plasticity, as well as determinants of interspecific variation, remain potential avenues for further exploration.
Collapse
Affiliation(s)
- Kerianne M Wilson
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, CA, USA
- Department of Biology, Pomona College, Claremont, CA, USA
| | - April M Arquilla
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, CA, USA
| | - Wendy Saltzman
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, CA, USA
- Neuroscience Graduate Program, University of California, Riverside, CA, USA
| |
Collapse
|
5
|
Beeson ALS, Meitzen J. Estrous cycle impacts on dendritic spine plasticity in rat nucleus accumbens core and shell and caudate-putamen. J Comp Neurol 2023; 531:759-774. [PMID: 36756791 PMCID: PMC10994586 DOI: 10.1002/cne.25460] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/22/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
An important factor that can modulate neuron properties is sex-specific hormone fluctuations, including the human menstrual cycle and rat estrous cycle in adult females. Considering the striatal brain regions, the nucleus accumbens (NAc) core, NAc shell, and caudate-putamen (CPu), the estrous cycle has previously been shown to impact relevant behaviors and disorders, neuromodulator action, and medium spiny neuron (MSN) electrophysiology. Whether the estrous cycle impacts MSN dendritic spine attributes has not yet been examined, even though MSN spines and glutamatergic synapse properties are sensitive to exogenously applied estradiol. Thus, we hypothesized that MSN dendritic spine attributes would differ by estrous cycle phase. To test this hypothesis, brains from adult male rats and female rats in diestrus, proestrus AM, proestrus PM, and estrus were processed for Rapid Golgi-Cox staining. MSN dendritic spine density, size, and type were analyzed in the NAc core, NAc shell, and CPu. Overall spine size differed across estrous cycle phases in female NAc core and NAc shell, and spine length differed across estrous cycle phase in NAc shell and CPu. Consistent with previous work, dendritic spine density was increased in the NAc core compared to the NAc shell and CPu, independent of sex and estrous cycle. Spine attributes in all striatal regions did not differ by sex when estrous cycle was disregarded. These results indicate, for the first time, that estrous cycle phase impacts dendritic spine plasticity in striatal regions, providing a neuroanatomical avenue by which sex-specific hormone fluctuations can impact striatal function and disorders.
Collapse
Affiliation(s)
- Anna LS Beeson
- Department of Biological Sciences, NC State University, Raleigh, USA
- Graduate Program in Biology, NC State University, Raleigh, USA
| | - John Meitzen
- Department of Biological Sciences, NC State University, Raleigh, USA
- Comparative Medicine Institute, NC State University, Raleigh, USA
- Center for Human Health and the Environment, NC State University, Raleigh, USA
| |
Collapse
|
6
|
Schecter RW, Jensen CM, Gavornik JP. Sex and estrous cycle affect experience-dependent plasticity in mouse primary visual cortex. PLoS One 2023; 18:e0282349. [PMID: 37068089 PMCID: PMC10109517 DOI: 10.1371/journal.pone.0282349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/13/2023] [Indexed: 04/18/2023] Open
Abstract
Sex hormones can affect cellular physiology and modulate synaptic plasticity, but it is not always clear whether or how sex-dependent differences identified in vitro express themselves as functional dimorphisms in the brain. Historically, most experimental neuroscience has been conducted using only male animals and the literature is largely mute about whether including female mice in will introduce variability due to inherent sex differences or endogenous estrous cycles. Though this is beginning to change following an NIH directive that sex should be included as a factor in vertebrate research, the lack of information raises practical issues around how to design experimental controls and apply existing knowledge to more heterogeneous populations. Various lines of research suggest that visual processing can be affected by sex and estrous cycle stage. For these reasons, we performed a series of in vivo electrophysiological experiments to characterize baseline visual function and experience-dependent plasticity in the primary visual cortex (V1) of male and female mice. We find that sex and estrous stage have no statistically significant effect on baseline acuity measurements, but that both sex and estrous stage have can modulate two mechanistically distinct forms of experience dependent cortical plasticity. We also demonstrate that resulting variability can be largely controlled with appropriate normalizations. These findings suggest that V1 plasticity can be used for mechanistic studies focusing on how sex hormones effect experience dependent plasticity in the mammalian cortex.
Collapse
Affiliation(s)
- Rachel W. Schecter
- Center for Systems Neuroscience, Biology Department, Boston University, Boston, MA, United States of America
| | - Cambria M. Jensen
- Center for Systems Neuroscience, Biology Department, Boston University, Boston, MA, United States of America
| | - Jeffrey P. Gavornik
- Center for Systems Neuroscience, Biology Department, Boston University, Boston, MA, United States of America
| |
Collapse
|
7
|
Pletzer B, Winkler-Crepaz K, Hillerer K. Progesterone and contraceptive progestin actions on the brain: A systematic review of animal studies and comparison to human neuroimaging studies. Front Neuroendocrinol 2023; 69:101060. [PMID: 36758768 DOI: 10.1016/j.yfrne.2023.101060] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 01/25/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
In this review we systematically summarize the effects of progesterone and synthetic progestins on neurogenesis, synaptogenesis, myelination and six neurotransmitter systems. Several parallels between progesterone and older generation progestin actions emerged, suggesting actions via progesterone receptors. However, existing results suggest a general lack of knowledge regarding the effects of currently used progestins in hormonal contraception regarding these cellular and molecular brain parameters. Human neuroimaging studies were reviewed with a focus on randomized placebo-controlled trials and cross-sectional studies controlling for progestin type. The prefrontal cortex, amygdala, salience network and hippocampus were identified as regions of interest for future preclinical studies. This review proposes a series of experiments to elucidate the cellular and molecular actions of contraceptive progestins in these areas and link these actions to behavioral markers of emotional and cognitive functioning. Emotional effects of contraceptive progestins appear to be related to 1) alterations in the serotonergic system, 2) direct/indirect modulations of inhibitory GABA-ergic signalling via effects on the allopregnanolone content of the brain, which differ between androgenic and anti-androgenic progestins. Cognitive effects of combined oral contraceptives appear to depend on the ethinylestradiol dose.
Collapse
Affiliation(s)
- Belinda Pletzer
- Department of Psychology & Centre for Cognitive Neuroscience, Paris-Lodron-University Salzburg, Salzburg Austria.
| | | | - Katharina Hillerer
- Department of Gynaecology & Obstetrics, Private Medical University, Salzburg, Austria
| |
Collapse
|
8
|
Velasco ER, Florido A, Perez-Caballero L, Marin I, Andero R. The Impacts of Sex Differences and Sex Hormones on Fear Extinction. Curr Top Behav Neurosci 2023; 64:105-132. [PMID: 37528309 DOI: 10.1007/7854_2023_426] [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] [Indexed: 08/03/2023]
Abstract
Fear extinction memories are strongly modulated by sex and hormonal status, but the exact mechanisms are still being discovered. In humans, there are some basal and task-related features in which male and female individuals differ in fear conditioning paradigms. However, analyses considering the effects of sex hormones demonstrate a role for estradiol in fear extinction memory consolidation. Translational studies are taking advantage of the convergent findings between species to understand the brain structures implicated. Nevertheless, the human brain is complex and the transfer of these findings into the clinics remains a challenge. The promising advances in the field together with the standardization of fear extinction methodologies in humans will benefit the design of new personalized therapies.
Collapse
Affiliation(s)
- Eric Raul Velasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antonio Florido
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Perez-Caballero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ignacio Marin
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Raul Andero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- ICREA, Pg. Lluís Companys 23, Barcelona, Spain.
| |
Collapse
|
9
|
Frankfurt M, Nassrallah Z, Luine V. Steroid Hormone Interaction with Dendritic Spines: Implications for Neuropsychiatric Disease. ADVANCES IN NEUROBIOLOGY 2023; 34:349-366. [PMID: 37962800 DOI: 10.1007/978-3-031-36159-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dendritic spines, key sites for neural plasticity, are influenced by gonadal steroids. In this chapter, we review the effects of gonadal steroids on dendritic spine density in areas important to cognitive function, the hippocampus, and prefrontal cortex. Most of these animal model studies investigated the effects of estrogen in females, but we also include more recent data on androgen effects in both males and females. The underlying genomic and non-genomic mechanisms related to gonadal steroid-induced spinogenesis are also reviewed. Subsequently, we discuss possible reasons for the observed sex differences in many neuropsychiatric diseases, which appear to be caused, in part, by aberrant synaptic connections that may involve dendritic spine pathology. Overall, knowledge concerning the regulation of dendritic spines by gonadal hormones has grown since the initial discoveries in the 1990s, and current research points to a potential role for aberrant spine functioning in many neuropsychiatric disorders.
Collapse
Affiliation(s)
- Maya Frankfurt
- Hofstra Northwell School of Nursing and Physician Assistant Studies, Hempstead, NY, USA.
| | - Zeinab Nassrallah
- Department of Science Education Zucker School of Medicine, 500 Hofstra University, Hempstead, NY, USA
| | - Victoria Luine
- Department of Psychology, Hunter College, New York, NY, USA
| |
Collapse
|
10
|
Hilz EN, Lee HJ. Estradiol and progesterone in female reward-learning, addiction, and therapeutic interventions. Front Neuroendocrinol 2023; 68:101043. [PMID: 36356909 DOI: 10.1016/j.yfrne.2022.101043] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/24/2022] [Accepted: 10/28/2022] [Indexed: 11/09/2022]
Abstract
Sex steroid hormones like estradiol (E2) and progesterone (P4) guide the sexual organization and activation of the developing brain and control female reproductive behavior throughout the lifecycle; importantly, these hormones modulate functional activity of not just the endocrine system, but most of the nervous system including the brain reward system. The effects of E2 and P4 can be seen in the processing of and memory for rewarding stimuli and in the development of compulsive reward-seeking behaviors like those seen in substance use disorders. Women are at increased risk of developing substance use disorders; however, the origins of this sex difference are not well understood and therapeutic interventions targeting ovarian hormones have produced conflicting results. This article reviews the contribution of the E2 and P4 in females to functional modulation of the brain reward system, their possible roles in origins of addiction vulnerability, and the development and treatment of compulsive reward-seeking behaviors.
Collapse
Affiliation(s)
- Emily N Hilz
- The University of Texas at Austin, Department of Pharmacology, USA.
| | - Hongjoo J Lee
- The University of Texas at Austin, Department of Psychology, USA; The University of Texas at Austin, Institute for Neuroscience, USA
| |
Collapse
|
11
|
Shanmugan S, Seidlitz J, Cui Z, Adebimpe A, Bassett DS, Bertolero MA, Davatzikos C, Fair DA, Gur RE, Gur RC, Larsen B, Li H, Pines A, Raznahan A, Roalf DR, Shinohara RT, Vogel J, Wolf DH, Fan Y, Alexander-Bloch A, Satterthwaite TD. Sex differences in the functional topography of association networks in youth. Proc Natl Acad Sci U S A 2022; 119:e2110416119. [PMID: 35939696 PMCID: PMC9388107 DOI: 10.1073/pnas.2110416119] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/15/2022] [Indexed: 01/16/2023] Open
Abstract
Prior work has shown that there is substantial interindividual variation in the spatial distribution of functional networks across the cerebral cortex, or functional topography. However, it remains unknown whether there are sex differences in the topography of individualized networks in youth. Here, we leveraged an advanced machine learning method (sparsity-regularized non-negative matrix factorization) to define individualized functional networks in 693 youth (ages 8 to 23 y) who underwent functional MRI as part of the Philadelphia Neurodevelopmental Cohort. Multivariate pattern analysis using support vector machines classified participant sex based on functional topography with 82.9% accuracy (P < 0.0001). Brain regions most effective in classifying participant sex belonged to association networks, including the ventral attention, default mode, and frontoparietal networks. Mass univariate analyses using generalized additive models with penalized splines provided convergent results. Furthermore, transcriptomic data from the Allen Human Brain Atlas revealed that sex differences in multivariate patterns of functional topography were spatially correlated with the expression of genes on the X chromosome. These results highlight the role of sex as a biological variable in shaping functional topography.
Collapse
Affiliation(s)
- Sheila Shanmugan
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Jakob Seidlitz
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Zaixu Cui
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Chinese Institute for Brain Research, Beijing,102206, China
| | - Azeez Adebimpe
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Danielle S. Bassett
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104
- Santa Fe Institute, Santa Fe, NM 87501
| | - Maxwell A. Bertolero
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Christos Davatzikos
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Damien A. Fair
- Department of Behavioral Neuroscience, Department of Psychiatry, Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR 97239
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Hongming Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Adam Pines
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Armin Raznahan
- Section on Developmental Neurogenomics Unit, Intramural Research Program, National Institutes of Mental Health, Bethesda, MD 20892
| | - David R. Roalf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Russell T. Shinohara
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104
| | - Jacob Vogel
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Daniel H. Wolf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Yong Fan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Aaron Alexander-Bloch
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Theodore D. Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
12
|
Oppong-Gyebi A, Metzger D, Vann PH, Yockey RA, Sumien N, Schreihofer DA. Dietary genistein and 17β-estradiol implants differentially influence locomotor and cognitive functions following transient focal ischemia in middle-aged ovariectomized rats at different lengths of estrogen deprivation. Horm Behav 2022; 144:105201. [PMID: 35653830 DOI: 10.1016/j.yhbeh.2022.105201] [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: 11/05/2021] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/04/2022]
Abstract
Genistein possesses estrogenic activity and has been considered a potential replacement for estrogen replacement therapy after menopause. In the current study, we investigated the neuroprotective effects of dietary genistein at varied lengths of estrogen deprivation in middle-aged ovariectomized Sprague-Dawley rats under ischemic conditions. Two weeks of treatment with dietary genistein at 42 mg/kg but not 17β-estradiol implants improved cognitive flexibility (Morris water maze test) after short-term estrogen deprivation (2 weeks) but not long-term estrogen deprivation (12 weeks). 17β-estradiol implants but not dietary genistein improved locomotor asymmetry (cylinder test) after long-term but not short-term estrogen deprivation. Dietary genistein but not 17β-estradiol implant improved early phase motor learning (rotarod test) after long-term estrogen deprivation. Neither 17β-estradiol implant nor dietary genistein reduced infarct size after either short-term or long-term estrogen deprivation. Genistein, however, reduced ionized calcium-binding adaptor molecule-1 (Iba1) expression, a marker of brain inflammation, at the ipsilateral side of stroke injury after short-term but not long-term estrogen deprivation. This study suggests that the neuroprotective effects of dietary genistein on motor and cognitive functions are distinctly influenced by the length of estrogen deprivation following focal ischemia. SIGNIFICANCE: There is an increasing postmenopausal population opting for homeopathic medicines for the management of menopausal symptoms due to the perceived distrust in estrogen use as hormone replacement. Basic and clinical studies support the notion that early, but not delayed, hormone replacement after menopause is beneficial. Furthermore, evidence suggests that delaying hormone replacement augments the detrimental, rather than the beneficial effects of estrogens. Because of the active consideration of soy isoflavones including genistein as alternatives to estrogen replacement, it is necessary to understand the ramifications of soy isoflavones use when their administration is begun at various times after menopause.
Collapse
Affiliation(s)
- Anthony Oppong-Gyebi
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Daniel Metzger
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Philip H Vann
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - R Andrew Yockey
- Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Derek A Schreihofer
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA; Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
13
|
de Souza RFL, Mendes TMAS, Lima LABDA, Brandão DS, Laplagne DA, de Sousa MBC. Effect of the Menstrual Cycle on Electroencephalogram Alpha and Beta Bands During Motor Imagery and Action Observation. Front Hum Neurosci 2022; 16:878887. [PMID: 35601901 PMCID: PMC9119141 DOI: 10.3389/fnhum.2022.878887] [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: 02/18/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Female sex steroids (FSS) can affect the motor system, modulating motor cortex excitability as well as performance in dexterity and coordination tasks. However, it has not yet been explored whether FSS affects the cognitive components of motor behavior. Mu is a sensorimotor rhythm observed by electroencephalography (EEG) in alpha (8–12 Hz) and beta (15–30 Hz) frequency bands in practices such as motor imagery (MI) and action observation (AO). This rhythm represents a window for studying the activity of neural circuits involved in motor cognition. Herein we investigated whether the alpha-mu and beta-mu power in the sensorimotor region (C3 and C4, hypothesis-driven approach) and the alpha and beta power over frontal, parietal, and occipital regions (data-driven approach) are modulated differently in the menstrual, follicular, and luteal phases of menstrual cycles in right-handed dominant women. To do so, these women underwent MI and AO in the three menstrual cycle phases. The spectral activity of the cortical regions for the alpha and beta bands were compared between phases of the menstrual cycle and a correlation analysis was also performed in relation to estrogen and progesterone levels. For the hypothesis-based approach, beta-mu event-related desynchronization (ERD) was significantly stronger in the C3 channel in the follicular phase than in the menstrual and luteal phases. For the data-driven approach, beta ERD during MI was higher in the follicular phase than in the menstrual and luteal phases in the frontal region. These findings suggest the effect of FSS on executive movement control. No effect of menstrual cycle phases was observed in cortical areas investigated during OA, but alpha and beta bands correlated positively with the follicular phase plasma estradiol level. Thus, the attenuation of alpha and beta bands referring to mirror neuron activities appears to be associated with inhibition of cortical activity when estradiol levels are lower, improving cognitive processing of motor action.
Collapse
Affiliation(s)
- Rafaela Faustino Lacerda de Souza
- Behavioral Endocrinology Laboratory, Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
- *Correspondence: Rafaela Faustino Lacerda de Souza,
| | | | | | - Daniel Soares Brandão
- Electroencephalography Laboratory, Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Diego Andrés Laplagne
- Behavioral Neurophysiology, Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Maria Bernardete Cordeiro de Sousa
- Behavioral Endocrinology Laboratory, Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
- Graduate Program in Psychobiology, Federal University of Rio Grande do Norte, Rio Grande do Norte, Brazil
- Maria Bernardete Cordeiro de Sousa,
| |
Collapse
|
14
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
15
|
Handley EE, Reale LA, Chuckowree JA, Dyer MS, Barnett GL, Clark CM, Bennett W, Dickson TC, Blizzard CA. Estrogen Enhances Dendrite Spine Function and Recovers Deficits in Neuroplasticity in the prpTDP-43A315T Mouse Model of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2022; 59:2962-2976. [PMID: 35249200 PMCID: PMC9016039 DOI: 10.1007/s12035-022-02742-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 01/09/2022] [Indexed: 10/31/2022]
Abstract
AbstractAmyotrophic lateral sclerosis (ALS) attacks the corticomotor system, with motor cortex function affected early in disease. Younger females have a lower relative risk of succumbing to ALS than males and older females, implicating a role for female sex hormones in disease progression. However, the mechanisms driving this dimorphic incidence are still largely unknown. We endeavoured to determine if estrogen mitigates disease progression and pathogenesis, focussing upon the dendritic spine as a site of action. Using two-photon live imaging we identify, in the prpTDP-43A315T mouse model of ALS, that dendritic spines in the male motor cortex have a reduced capacity for remodelling than their wild-type controls. In contrast, females show higher capacity for remodelling, with peak plasticity corresponding to highest estrogen levels during the estrous cycle. Estrogen manipulation through ovariectomies and estrogen replacement with 17β estradiol in vivo was found to significantly alter spine density and mitigate disease severity. Collectively, these findings reveal that synpatic plasticity is reduced in ALS, which can be amelioriated with estrogen, in conjuction with improved disease outcomes.
Collapse
|
16
|
Navakkode S, Gaunt JR, Pavon MV, Bansal VA, Abraham RP, Chong YS, Ch'ng TH, Sajikumar S. Sex-specific accelerated decay in time/activity-dependent plasticity and associative memory in an animal model of Alzheimer's disease. Aging Cell 2021; 20:e13502. [PMID: 34796608 PMCID: PMC8672784 DOI: 10.1111/acel.13502] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/02/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical studies have shown that female brains are more predisposed to neurodegenerative diseases such as Alzheimer's disease (AD), but the cellular and molecular mechanisms behind this disparity remain unknown. In several mouse models of AD, synaptic plasticity dysfunction is an early event and appears before significant accumulation of amyloid plaques and neuronal degeneration. However, it is unclear whether sexual dimorphism at the synaptic level contributes to the higher risk and prevalence of AD in females. Our studies on APP/PS1 (APPSwe/PS1dE9) mouse model show that AD impacts hippocampal long‐term plasticity in a sex‐specific manner. Long‐term potentiation (LTP) induced by strong tetanic stimulation (STET), theta burst stimulation (TBS) and population spike timing‐dependent plasticity (pSTDP) show a faster decay in AD females compared with age‐matched AD males. In addition, behavioural tagging (BT), a model of associative memory, is specifically impaired in AD females with a faster decay in memory compared with males. Together with the plasticity and behavioural data, we also observed an upregulation of neuroinflammatory markers, along with downregulation of transcripts that regulate cellular processes associated with synaptic plasticity and memory in females. Immunohistochemistry of AD brains confirms that female APP/PS1 mice carry a higher amyloid plaque burden and have enhanced microglial activation compared with male APP/PS1 mice. Their presence in the diseased mice also suggests a link between the impairment of LTP and the upregulation of the inflammatory response. Overall, our data show that synaptic plasticity and associative memory impairments are more prominent in females and this might account for the faster progression of AD in females.
Collapse
Affiliation(s)
- Sheeja Navakkode
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Department of Physiology National University of Singapore Singapore Singapore
| | - Jessica Ruth Gaunt
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Maria Vazquez Pavon
- Department of Physiology National University of Singapore Singapore Singapore
| | | | - Riya Prasad Abraham
- Department of Physiology National University of Singapore Singapore Singapore
| | - Yee Song Chong
- Department of Physiology National University of Singapore Singapore Singapore
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- School of Biological Science Nanyang Technological University Singapore Singapore
| | - Sreedharan Sajikumar
- Department of Physiology National University of Singapore Singapore Singapore
- Healthy Longevity Translational Research Programme Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Life Sciences Institute Neurobiology Programme National University of Singapore Singapore Singapore
| |
Collapse
|
17
|
Changes in dendritic arborization related to the estrous cycle in pyramidal neurons of layer V of the motor cortex. J Chem Neuroanat 2021; 119:102042. [PMID: 34800658 DOI: 10.1016/j.jchemneu.2021.102042] [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: 03/19/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022]
Abstract
Many studies on neuronal plasticity have been conducted in the hippocampus and sensory cortices. In female rats in the estrus phase, when there is a low concentration of estradiol in the blood, there is a reduction in the dendritic spine density of CA1 neurons, while an increase in dendritic spines has been observed during metestrus, when progesterone levels are high. In comparison with the hippocampus, less information is known about dendritic remodeling of the motor cortex. Thus, the objective of the present study was to evaluate the neuronal morphology of pyramidal cells of layer V of the motor cortex in each phase of the estrous cycle. For this, we used Long-Evans strain rats and formed 4 experimental groups according to the phase of the estrous cycle at the moment of sacrifice: proestrus, estrus, metestrus, or diestrus. All animals were gently monitored regarding the expression of one estrous cycle in order to determine the regularity of the cycle. We obtained the brains in order to evaluate the neuronal morphology of neurons of layer V of the primary motor cortex following the Golgi-Cox method and Sholl analysis. Our results show that the dendritic arborization of neurons of rats sacrificed in the metestrus phase is reduced compared to the other phases of the estrous cycle. However, we did not find changes in dendritic spine density between experimental groups. When comparing our results with previous data, we can suggest that estrogens and progesterone differentially promote plasticity events in pyramidal neurons between different brain regions.
Collapse
|
18
|
Oppong-Gyebi A, Metzger D, Doan T, Han J, Vann PH, Yockey RA, Sumien N, Schreihofer DA. Long-term hypogonadism diminishes the neuroprotective effects of dietary genistein in young adult ovariectomized rats after transient focal ischemia. J Neurosci Res 2021; 100:598-619. [PMID: 34713481 DOI: 10.1002/jnr.24981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 08/19/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023]
Abstract
Increasing age disproportionately increases the risk of stroke among women compared to men of similar age, especially after menopause. One of the reasons for this observation is a sharp drop in circulating estrogens. However, the timing of initiation of estrogen replacement after menopause is associated with mixed beneficial and detrimental effects, hence contributing to widespread mistrust of estrogen use. Agents including soy isoflavones are being assessed as viable alternatives to estrogen therapy. In this study, we hypothesized that the neuroprotective effects of genistein, a soy isoflavone are less sensitive to the length of hypogonadism in young adult ovariectomized rats following cerebral ischemia. We expected that long-term hypogonadism will worsen motor and cognitive function, increase post-stroke inflammation with no effect on the neuroprotection of genistein. We compared the effect of treatment with dietary genistein (GEN) on short-term (2 weeks) and long-term hypogonadism (12 weeks) in young adult ovariectomized Sprague-Dawley rats on sensorimotor function, cognition and inflammation after focal ischemia. Dorsal Silastic implant of 17β-estradiol (E2) was used as a control for hormone therapy. Long-term hypogonadism stroked rats performed worse than the short-term hypogonadism stroked rats on the motor and cognitive function tests. GEN did not improve neurological assessment and motor learning after either short-term or long-term hypogonadism. GEN improved cognitive flexibility after short-term hypogonadism but not after the long-term. Both GEN and E2 reduced tissue loss after short-term hypogonadism and reduced GFAP expression at the contralateral side of ischemia after long-term hypogonadism. The length of hypogonadism may differentially influence the neuroprotective effects of both GEN and E2 on the motor and cognitive functions in young adult rats.
Collapse
Affiliation(s)
- Anthony Oppong-Gyebi
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA.,Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Daniel Metzger
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA.,Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Trinh Doan
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Jordan Han
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Phillip H Vann
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA.,Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - R Andrew Yockey
- Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA.,Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Derek A Schreihofer
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA.,Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas, USA
| |
Collapse
|
19
|
Estrogenic hormones receptors in Alzheimer's disease. Mol Biol Rep 2021; 48:7517-7526. [PMID: 34657250 DOI: 10.1007/s11033-021-06792-1] [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: 07/21/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023]
Abstract
Estrogens are hormones that play a critical role during development and growth for the adequate functioning of the reproductive system of women, as well as for maintaining bones, metabolism, and cognition. During menopause, the levels of estrogens are decreased, altering their signaling mediated by their intracellular receptors such as estrogen receptor alpha and beta (ERα and ERβ), and G protein-coupled estrogen receptor (GPER). In the brain, the reduction of molecular pathways mediated by estrogenic receptors seems to favor the progression of Alzheimer's disease (AD) in postmenopausal women. In this review, we investigate the participation of estrogen receptors in AD in women during aging.
Collapse
|
20
|
Fahrenkopf A, Li G, Wood RI, Wagner CK. Developmental exposure to the synthetic progestin, 17α-hydroxyprogesterone caproate, disrupts the mesocortical serotonin pathway and alters impulsive decision-making in rats. Dev Neurobiol 2021; 81:763-773. [PMID: 34318625 PMCID: PMC8440456 DOI: 10.1002/dneu.22847] [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] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 01/25/2023]
Abstract
The synthetic progestin, 17α-hydroxyprogesterone caproate (17-OHPC), is administered to women at risk for preterm birth during a critical period of fetal development for mesocortical pathways. Yet, little information is available regarding the potential effects of 17-OHPC on the developing fetal brain. In rat models, the mesocortical serotonin pathway is sensitive to progestins. Progesterone receptor (PR) is expressed in layer 3 pyramidal neurons of medial prefrontal cortex (mPFC) and in serotonergic neurons of the dorsal raphe. The present study tested the hypothesis that exposure to 17-OHPC during development disrupts serotonergic innervation of the mPFC in adolescence and impairs behavior mediated by this pathway in adulthood. Administration of 17-OHPC from postnatal days 1-14 decreased the density of SERT-ir fibers within superficial and deep layers and decreased the density of synaptophysin-ir boutons in all layers of prelimbic mPFC at postnatal day 28. In addition, rats exposed to 17-OHPC during development were less likely to make impulsive choices in the Delay Discounting task, choosing the larger, delayed reward more often than controls at moderate delay times. Interestingly, 17-OHPC exposed rats were more likely to fail to make any choice (i.e., increased omissions) compared to controls at longer delays, suggesting disruptions in decision-making. These results suggest that further investigation is warranted in the clinical use of 17-OHPC to better inform a risk/benefit analysis of progestin use in pregnancy.
Collapse
Affiliation(s)
- Allyssa Fahrenkopf
- Psychogenics Inc. Paramus, NJ USA
- Department of Psychology & Center for Neuroscience Research, University at Albany, Albany, NY USA
| | - Grace Li
- Department of Integrative Anatomical Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Ruth I. Wood
- Department of Integrative Anatomical Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Christine K. Wagner
- Department of Psychology & Center for Neuroscience Research, University at Albany, Albany, NY USA
| |
Collapse
|
21
|
Zhang M, Spencer HF, Berman RY, Radford KD, Choi KH. Effects of subanesthetic intravenous ketamine infusion on neuroplasticity-related proteins in male and female Sprague-Dawley rats. IBRO Neurosci Rep 2021; 11:42-51. [PMID: 34286313 PMCID: PMC8273220 DOI: 10.1016/j.ibneur.2021.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 10/30/2022] Open
Abstract
Although ketamine, a multimodal dissociative anesthetic, is frequently used for analgesia and treatment-resistant major depression, molecular mechanisms of ketamine remain unclear. Specifically, differences in the effects of ketamine on neuroplasticity-related proteins in the brains of males and females need further investigation. In the current study, adult male and female Sprague-Dawley rats with an indwelling jugular venous catheter received an intravenous ketamine infusion (0, 10, or 40 mg/kg, 2-h), starting with a 2 mg/kg bolus for ketamine groups. Spontaneous locomotor activity was monitored by infrared photobeams during the infusion. Two hours after the infusion, brain tissue was dissected to obtain the medial prefrontal cortex (mPFC), hippocampus including the CA1, CA3, and dentate gyrus, and amygdala followed by Western blot analyses of a transcription factor (c-Fos), brain-derived neurotrophic factor (BDNF), and phosphorylated extracellular signal-regulated kinase (pERK). The 10 mg/kg ketamine infusion suppressed locomotor activity in male and female rats while the 40 mg/kg infusion stimulated activity only in female rats. In the mPFC, 10 mg/kg ketamine reduced pERK levels in male rats while 40 mg/kg ketamine increased c-Fos levels in male and female rats. Female rats in proestrus/estrus phases showed greater ketamine-induced c-Fos elevation as compared to those in diestrus phase. In the amygdala, 10 and 40 mg/kg ketamine increased c-Fos levels in female, but not male, rats. In the hippocampus, 10 mg/kg ketamine reduced BDNF levels in male, but not female, rats. Taken together, the current data suggest that subanesthetic doses of intravenous ketamine infusions produce differences in neuroplasticity-related proteins in the brains of male and female rats.
Collapse
Affiliation(s)
- Michael Zhang
- Department of Psychiatry, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.,Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Haley F Spencer
- Program in Neuroscience, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Rina Y Berman
- Department of Psychiatry, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.,Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Kennett D Radford
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Kwang H Choi
- Department of Psychiatry, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.,Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.,Program in Neuroscience, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.,Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
22
|
Meeker TJ, Veldhuijzen DS, Keaser ML, Gullapalli RP, Greenspan JD. Menstrual Cycle Variations in Gray Matter Volume, White Matter Volume and Functional Connectivity: Critical Impact on Parietal Lobe. Front Neurosci 2020; 14:594588. [PMID: 33414702 PMCID: PMC7783210 DOI: 10.3389/fnins.2020.594588] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
The role of gonadal hormones in neural plasticity remains unclear. This study aimed to examine the effects of naturally fluctuating hormone levels over the menstrual cycle in healthy females. Gray matter, functional connectivity (FC) and white matter changes over the cycle were assessed by using functional magnetic resonance imaging (fMRI), resting state fMRI, and structural MRIs, respectively, and associated with serum gonadal hormone levels. Moreover, electrocutaneous sensitivity was evaluated in 14 women in four phases of their menstrual cycle (menstrual, follicular, ovulatory, and luteal). Electrocutaneous sensitivity was greater during follicular compared to menstrual phase. Additionally, pain unpleasantness was lower in follicular phase than other phases while pain intensity ratings did not change over the cycle. Significant variations in cycle phase effects on gray matter volume were found in the left inferior parietal lobule (IPL) using voxel-based morphometry. Subsequent Freesurfer analysis revealed greater thickness of left IPL during the menstrual phase when compared to other phases. Also, white matter volume fluctuated across phases in left IPL. Blood estradiol was positively correlated with white matter volume both in left parietal cortex and whole cortex. Seed-driven FC between left IPL and right secondary visual cortex was enhanced during ovulatory phase. A seed placed in right IPL revealed enhanced FC between left and right IPL during the ovulatory phase. Additionally, we found that somatosensory cortical gray matter was thinner during follicular compared to menstrual phase. We discuss these results in the context of likely evolutionary pressures selecting for enhanced perceptual sensitivity across modalities specifically during ovulation.
Collapse
Affiliation(s)
- Timothy J. Meeker
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
- Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, United States
| | - Dieuwke S. Veldhuijzen
- Institute of Psychology, Health, Medical and Neuropsychology Unit, Leiden University, Leiden, Netherlands
- Leiden Institute for Brain and Cognition, Leiden, Netherlands
| | - Michael L. Keaser
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
- Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, United States
| | - Rao P. Gullapalli
- Department of Diagnostic Radiology and Nuclear Imaging, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Joel D. Greenspan
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
- Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, United States
| |
Collapse
|
23
|
McCarthy M, Raval AP. The peri-menopause in a woman's life: a systemic inflammatory phase that enables later neurodegenerative disease. J Neuroinflammation 2020; 17:317. [PMID: 33097048 PMCID: PMC7585188 DOI: 10.1186/s12974-020-01998-9] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023] Open
Abstract
The peri-menopause or menopausal transition—the time period that surrounds the final years of a woman’s reproductive life—is associated with profound reproductive and hormonal changes in a woman’s body and exponentially increases a woman’s risk of cerebral ischemia and Alzheimer’s disease. Although our understanding of the exact timeline or definition of peri-menopause is limited, it is clear that there are two stages to the peri-menopause. These are the early menopausal transition, where menstrual cycles are mostly regular, with relatively few interruptions, and the late transition, where amenorrhea becomes more prolonged and lasts for at least 60 days, up to the final menstrual period. Emerging evidence is showing that peri-menopause is pro-inflammatory and disrupts estrogen-regulated neurological systems. Estrogen is a master regulator that functions through a network of estrogen receptors subtypes alpha (ER-α) and beta (ER-β). Estrogen receptor-beta has been shown to regulate a key component of the innate immune response known as the inflammasome, and it also is involved in regulation of neuronal mitochondrial function. This review will present an overview of the menopausal transition as an inflammatory event, with associated systemic and central nervous system inflammation, plus regulation of the innate immune response by ER-β-mediated mechanisms.
Collapse
Affiliation(s)
- Micheline McCarthy
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA. .,Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
24
|
Acharya KD, Nettles SA, Lichti CF, Warre-Cornish K, Polit LD, Srivastava DP, Denner L, Tetel MJ. Dopamine-induced interactions of female mouse hypothalamic proteins with progestin receptor-A in the absence of hormone. J Neuroendocrinol 2020; 32:e12904. [PMID: 33000549 PMCID: PMC7591852 DOI: 10.1111/jne.12904] [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: 04/22/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 11/26/2022]
Abstract
Neural progestin receptors (PR) function in reproduction, neural development, neuroprotection, learning, memory and the anxiety response. In the absence of progestins, PR can be activated by dopamine (DA) in the rodent hypothalamus to elicit female sexual behaviour. The present study investigated mechanisms of DA activation of PR by testing the hypothesis that proteins from DA-treated hypothalami interact with PR in the absence of progestins. Ovariectomised, oestradiol-primed mice were infused with a D1-receptor agonist, SKF38393 (SKF), into the third ventricle 30 minutes prior to death. Proteins from SKF-treated hypothalami were pulled-down with glutathione S-transferase-tagged mouse PR-A or PR-B and the interactomes were analysed by mass spectrometry. The largest functional group to interact with PR-A in a DA-dependent manner was synaptic proteins. To test the hypothesis that DA activation of PR regulates synaptic proteins, we developed oestradiol-induced PR-expressing hypothalamic-like neurones derived from human-induced pluripotent stem cells (hiPSCs). Similar to progesterone (P4), SKF treatment of hiPSCs increased synapsin1/2 expression. This SKF-dependent effect was blocked by the PR antagonist RU486, suggesting that PR are necessary for this DA-induced increase. The second largest DA-dependent PR-A protein interactome comprised metabolic regulators involved in glucose metabolism, lipid synthesis and mitochondrial energy production. Interestingly, hypothalamic proteins interacted with PR-A, but not PR-B, in an SKF-dependent manner, suggesting that DA promotes the interaction of multiple hypothalamic proteins with PR-A. These in vivo and in vitro results indicate novel mechanisms by which DA can differentially activate PR isoforms in the absence of P4 and provide a better understanding of ligand-independent PR activation in reproductive, metabolic and mental health disorders in women.
Collapse
Affiliation(s)
| | | | - Cheryl F. Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110
| | - Katherine Warre-Cornish
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Lucia Dutan Polit
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Larry Denner
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Marc J. Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA 02481
| |
Collapse
|
25
|
Abstract
17β-Estradiol (E2) is a potent steroid hormone of both gonadal and neuronal origin that exerts profound effects on neuroplasticity in several brain regions. Dendritic spine and synapse formation and rearrangements are modulated and mediated by estrogens. In this chapter, we highlighted the essential background concerning the effects of E2 on synaptic rearrangements accompanied by synaptic plasticity in E2-sensitive brain regions that mediate learning and memory, i.e., cortex and hippocampus. We also address details of the molecular mechanisms underlying E2 regulation of spine dynamics. The proposed models of action of E2 overlaps with that for well-established synaptic modulators, such as adenosine. Thus, the possible synergistic effects of those two molecules in respect to synaptic rearrangement and plasticity were presented.
Collapse
|
26
|
Parker EM, Kindja NL, Cheetham CEJ, Sweet RA. Sex differences in dendritic spine density and morphology in auditory and visual cortices in adolescence and adulthood. Sci Rep 2020; 10:9442. [PMID: 32523006 PMCID: PMC7287134 DOI: 10.1038/s41598-020-65942-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/24/2020] [Indexed: 11/24/2022] Open
Abstract
Dendritic spines are small protrusions on dendrites that endow neurons with the ability to receive and transform synaptic input. Dendritic spine number and morphology are altered as a consequence of synaptic plasticity and circuit refinement during adolescence. Dendritic spine density (DSD) is significantly different based on sex in subcortical brain regions associated with the generation of sex-specific behaviors. It is largely unknown if sex differences in DSD exist in auditory and visual brain regions and if there are sex-specific changes in DSD in these regions that occur during adolescent development. We analyzed dendritic spines in 4-week-old (P28) and 12-week-old (P84) male and female mice and found that DSD is lower in female mice due in part to fewer short stubby, long stubby and short mushroom spines. We found striking layer-specific patterns including a significant age by layer interaction and significantly decreased DSD in layer 4 from P28 to P84. Together these data support the possibility of developmental sex differences in DSD in visual and auditory regions and provide evidence of layer-specific refinement of DSD over adolescent brain development.
Collapse
Affiliation(s)
- Emily M Parker
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, USA
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | - Nathan L Kindja
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | - Claire E J Cheetham
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, USA
- Center for the Neural Basis of Cognition, Pittsburgh, USA
| | - Robert A Sweet
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, USA.
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, USA.
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
27
|
Khaliulin I, Kartawy M, Amal H. Sex Differences in Biological Processes and Nitrergic Signaling in Mouse Brain. Biomedicines 2020; 8:biomedicines8050124. [PMID: 32429146 PMCID: PMC7277573 DOI: 10.3390/biomedicines8050124] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/06/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
Nitric oxide (NO) represents an important signaling molecule which modulates the functions of different organs, including the brain. S-nitrosylation (SNO), a post-translational modification that involves the binding of the NO group to a cysteine residue, is a key mechanism of nitrergic signaling. Most of the experimental studies are carried out on male animals. However, significant differences exist between males and females in the signaling mechanisms. To investigate the sex differences in the SNO-based regulation of biological functions and signaling pathways in the cortices of 6–8-weeks-old mice, we used the mass spectrometry technique, to identify S-nitrosylated proteins, followed by large-scale computational biology. This work revealed significant sex differences in the NO and SNO-related biological functions in the cortices of mice for the first-time. The study showed significant SNO-induced enrichment of the synaptic processes in female mice, but enhanced SNO-related cytoskeletal processes in the male mice. Proteins, which were S-nitrosylated in the cortices of mice of both groups, were more abundant in the female brain. Finally, we investigated the shared molecular processes that were found in both sexes. This study presents a mechanistic insight into the role of S-nitrosylation in both sexes and provides strong evidence of sex difference in many biological processes and signalling pathways, which will open future research directions on sex differences in neurological disorders.
Collapse
|
28
|
Hrynchak MV, Rierola M, Golovyashkina N, Penazzi L, Pump WC, David B, Sündermann F, Brandt R, Bakota L. Chronic Presence of Oligomeric Aβ Differentially Modulates Spine Parameters in the Hippocampus and Cortex of Mice With Low APP Transgene Expression. Front Synaptic Neurosci 2020; 12:16. [PMID: 32390822 PMCID: PMC7194154 DOI: 10.3389/fnsyn.2020.00016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/25/2020] [Indexed: 01/06/2023] Open
Abstract
Alzheimer’s disease is regarded as a synaptopathy with a long presymptomatic phase. Soluble, oligomeric amyloid-β (Aβ) is thought to play a causative role in this disease, which eventually leads to cognitive decline. However, most animal studies have employed mice expressing high levels of the Aβ precursor protein (APP) transgene to drive pathology. Here, to understand how the principal neurons in different brain regions cope with moderate, chronically present levels of Aβ, we employed transgenic mice expressing equal levels of mouse and human APP carrying a combination of three familial AD (FAD)-linked mutations (Swedish, Dutch, and London), that develop plaques only in old age. We analyzed dendritic spine parameters in hippocampal and cortical brain regions after targeted expression of EGFP to allow high-resolution imaging, followed by algorithm-based evaluation of mice of both sexes from adolescence to old age. We report that Aβ species gradually accumulated throughout the life of APPSDL mice, but not the oligomeric forms, and that the amount of membrane-associated oligomers decreased at the onset of plaque formation. We observed an age-dependent loss of thin spines under most conditions as an indicator of a loss of synaptic plasticity in older mice. We further found that hippocampal pyramidal neurons respond to increased Aβ levels by lowering spine density and shifting spine morphology, which reached significance in the CA1 subfield. In contrast, the spine density in cortical pyramidal neurons of APPSDL mice was unchanged. We also observed an increase in the protein levels of PSD-95 and Arc in the hippocampus and cortex, respectively. Our data demonstrated that increased concentrations of Aβ have diverse effects on dendritic spines in the brain and suggest that hippocampal and cortical neurons have different adaptive and compensatory capacity during their lifetime. Our data also indicated that spine morphology differs between sexes in a region-specific manner.
Collapse
Affiliation(s)
- Mariya V Hrynchak
- Department of Neurobiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Marina Rierola
- Department of Neurobiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Nataliya Golovyashkina
- Department of Neurobiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Lorène Penazzi
- Department of Neurobiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Wiebke C Pump
- Department of Neurobiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Bastian David
- Department of Neurobiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Frederik Sündermann
- Department of Neurobiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.,Center for Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany.,Institute of Cognitive Science, University of Osnabrück, Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
29
|
Havekes R, Aton SJ. Impacts of Sleep Loss versus Waking Experience on Brain Plasticity: Parallel or Orthogonal? Trends Neurosci 2020; 43:385-393. [PMID: 32459991 DOI: 10.1016/j.tins.2020.03.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 12/21/2022]
Abstract
Recent studies on the effects of sleep deprivation on synaptic plasticity have yielded discrepant results. Sleep deprivation studies using novelty exposure as a means to keep animals awake suggests that sleep (compared with wake) leads to widespread reductions in net synaptic strength. By contrast, sleep deprivation studies using approaches avoiding novelty-induced arousal (i.e., gentle handling) suggest that sleep can promote synaptic growth and strengthening. How can these discrepant findings be reconciled? Here, we discuss how varying methodologies for the experimental disruption of sleep (with differential introduction of novel experiences) could fundamentally alter the experimental outcome with regard to synaptic plasticity. Thus, data from experiments aimed at assessing the relative impact of sleep versus wake on the brain may instead reflect the quality of the waking experience itself. The highlighted work suggests that brain plasticity resulting from novel experiences versus wake per se has unique and distinct features.
Collapse
Affiliation(s)
- Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, Groningen, The Netherlands.
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, College of Literature, Sciences, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
30
|
Tsai ST, Chen SY, Lin SZ, Tseng GF. Rostral intralaminar thalamic deep brain stimulation ameliorates memory deficits and dendritic regression in β-amyloid-infused rats. Brain Struct Funct 2020; 225:751-761. [PMID: 32036422 DOI: 10.1007/s00429-020-02033-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 01/22/2020] [Indexed: 10/25/2022]
Abstract
Rostral intralaminar thalamic deep brain stimulation (ILN-DBS) has been shown to enhance attention and cognition through neuronal activation and brain plasticity. We examined whether rostral ILN-DBS can also attenuate memory deficits and impaired synaptic plasticity and protect glutamatergic transmission in the rat intraventricular β-amyloid (Aβ) infusion model of Alzheimer's disease (AD). Spatial memory was tested in the Morris water maze (MWM), while structural synaptic plasticity and glutamatergic transmission strength were estimated by measuring dendritic spine densities in dye-injected neurons and tissue expression levels of postsynaptic density protein 95 (PSD-95) in medial prefrontal cortex (mPFC) and hippocampus. All these assessments were compared among the naïve control rats, AD rats, and AD rats with ILN-DBS. We found that a single rostral ILN-DBS treatment significantly improved MWM performance and reversed PSD-95 expression reductions in the mPFC and hippocampal region of Aβ-infused rats. In addition, ILN-DBS preserved dendritic spine densities on mPFC and hippocampal pyramidal neurons. In fact, MWM performance, PSD-95 expression levels, and dendritic spine densities did not differ between naïve control and rostral ILN-DBS treatment groups, indicating near complete amelioration of Aβ-induced spatial memory impairments and dendritic regression. These findings suggest that the ILN is critical for modulating glutamatergic transmission, neural plasticity, and spatial memory functions through widespread effects on distributed brain regions. Further, these findings provide a rationale for examining the therapeutic efficacy of ILN-DBS in AD patients.
Collapse
Affiliation(s)
- Sheng-Tzung Tsai
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation/Tzu Chi University, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Shin-Yuan Chen
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation/Tzu Chi University, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation/Tzu Chi University, Hualien, Taiwan
| | - Guo-Fang Tseng
- Department of Anatomy, College of Medicine, Tzu-Chi University, No. 701, Section 3, Jhongyang Road, Hualien, 970, Taiwan.
| |
Collapse
|
31
|
Gildawie KR, Honeycutt JA, Brenhouse HC. Region-specific Effects of Maternal Separation on Perineuronal Net and Parvalbumin-expressing Interneuron Formation in Male and Female Rats. Neuroscience 2019; 428:23-37. [PMID: 31887358 DOI: 10.1016/j.neuroscience.2019.12.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/02/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022]
Abstract
Early life experiences play a vital role in contributing to healthy brain development. Adverse experiences have a lasting impact on the prefrontal cortex (PFC) and basolateral amygdala (BLA), brain regions associated with emotion regulation. Early life adversity via maternal separation (MS) has sex-specific effects on expression of parvalbumin (PV), which is expressed in fast-spiking GABAergic interneurons that are preferentially enwrapped by perineuronal nets (PNNs). Importantly, PNN formation coincides with the closure of developmental critical periods and regulates PV-expressing interneuron activity. Since aberrant PNN organization has been reported following adverse experiences in adolescent and adult rats, we investigated the impact of adversity early in life in the form of MS on the developing brain. Rat pups were separated from their dams for 4 h per day from postnatal day (P) 2-20. Tissue sections from juvenile (P20), adolescent (P40), and early adult (P70) animals containing the PFC and BLA were fluorescently stained to visualize Wisteria floribunda agglutinin+ PNNs and PV-expressing interneurons, and density and intensity was quantified. Our results confirm past reports that PFC PNNs form gradually throughout development; however, PNN density plateaus in adolescence, while intensity continues to increase into adulthood. Importantly, MS delays PNN formation in the prelimbic PFC and results in sex-specific aberrations in PNN structural integrity that do not appear until adulthood. The present findings reveal sex-, age-, and region-specific effects of early life adversity on PNN and PV maturation, implicating neuroplastic alterations following early life adversity that may be associated with sex differences in psychopathology and resilience.
Collapse
Affiliation(s)
- Kelsea R Gildawie
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA
| | - Jennifer A Honeycutt
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA
| | - Heather C Brenhouse
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
32
|
17α Estradiol promotes plasticity of spared inputs in the adult amblyopic visual cortex. Sci Rep 2019; 9:19040. [PMID: 31836739 PMCID: PMC6910995 DOI: 10.1038/s41598-019-55158-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 11/20/2019] [Indexed: 01/10/2023] Open
Abstract
The promotion of structural and functional plasticity by estrogens is a promising approach to enhance central nervous system function in the aged. However, how the sensitivity to estrogens is regulated across brain regions, age and experience is poorly understood. To ask if estradiol treatment impacts structural and functional plasticity in sensory cortices, we examined the acute effect of 17α-Estradiol in adult Long Evans rats following chronic monocular deprivation, a manipulation that reduces the strength and selectivity of deprived eye vision. Chronic monocular deprivation decreased thalamic input from the deprived eye to the binocular visual cortex and accelerated short-term depression of the deprived eye pathway, but did not change the density of excitatory synapses in primary visual cortex. Importantly, we found that the classical estrogen receptors ERα and ERβ were robustly expressed in the adult visual cortex, and that a single dose of 17α-Estradiol reduced the expression of the calcium-binding protein parvalbumin, decreased the integrity of the extracellular matrix and increased the size of excitatory postsynaptic densities. Furthermore, 17α-Estradiol enhanced experience-dependent plasticity in the amblyopic visual cortex, by promoting response potentiation of the pathway served by the non-deprived eye. The promotion of plasticity at synapses serving the non-deprived eye may reflect selectivity for synapses with an initially low probability of neurotransmitter release, and may inform strategies to remap spared inputs around a scotoma or a cortical infarct.
Collapse
|
33
|
Monciunskaite R, Malden L, Lukstaite I, Ruksenas O, Griksiene R. Do oral contraceptives modulate an ERP response to affective pictures? Biol Psychol 2019; 148:107767. [PMID: 31509765 DOI: 10.1016/j.biopsycho.2019.107767] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/23/2019] [Accepted: 09/06/2019] [Indexed: 12/22/2022]
Abstract
Indications exist that use of oral contraceptives affects women's socio-emotional behaviour, brain function and, cognitive abilities, but the information is still scarce and ambiguous. We aimed to examine affective processing of visual stimuli between oral contraceptive users (OC, n = 33) and naturally cycling women (NC, n = 37) using the event-related potential (ERP) method. The main findings are: (i) emotionally arousing stimuli elicited significantly enlarged late positive potential (LPP) amplitudes compared to neutral stimuli, (ii) anti-androgenic OC users demonstrated diminished brain reactivity to visual stimuli, and (iii) significantly blunted reaction to highly unpleasant images. In addition, a positive relationship between GFP evoked by the highly unpleasant and highly pleasant visual emotional stimuli and progesterone was observed in NC women, while OC users demonstrated a trend of negative relationship between GFP and progesterone level. These findings suggest possible modulations of affective processing of visual stimuli when hormonal contraceptives are used.
Collapse
Affiliation(s)
- R Monciunskaite
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| | - L Malden
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - I Lukstaite
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - O Ruksenas
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - R Griksiene
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
34
|
Kilpatrick LA, Holmberg M, Manzouri A, Savic I. Cross sex hormone treatment is linked with a reversal of cerebral patterns associated with gender dysphoria to the baseline of cisgender controls. Eur J Neurosci 2019; 50:3269-3281. [PMID: 30991464 DOI: 10.1111/ejn.14420] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/27/2019] [Accepted: 04/03/2019] [Indexed: 12/22/2022]
Abstract
Transgender persons experience incongruence between their gender identity and birth-assigned sex. The resulting gender dysphoria (GD), is frequently treated with cross-sex hormones. However, very little is known about how this treatment affects the brain of individuals with GD, nor do we know the neurobiology of GD. We recently suggested that disconnection of fronto-parietal networks involved in own-body self-referential processing could be a plausible mechanism, and that the anatomical correlate could be a thickening of the mesial prefrontal and precuneus cortex, which is unrelated to sex. Here, we investigate how cross-sex hormone treatment affects cerebral tissue in persons with GD, and how potential changes are related to self-body perception. Longitudinal MRI measurements of cortical thickness (Cth) were carried out in 40 transgender men (TrM), 24 transgender women (TrW) and 19 controls. Cth increased in the mesial temporal and insular cortices with testosterone treatment in TrM, whereas anti-androgen and oestrogen treatment in TrW caused widespread cortical thinning. However, after correction for treatment-related changes in total grey and white matter volumes (increase with testosterone; decrease with anti-androgen and oestrogen), significant Cth decreases were observed in the mesial prefrontal and parietal cortices, in both TrM and TrW (vs. controls) - regions showing greater pre-treatment Cth than in controls. The own body - self congruence ratings increased with treatment, and correlated with a left parietal cortical thinning. These data confirm our hypothesis that GD may be associated with specific anatomical features in own-body/self-processing circuits that reverse to the pattern of cisgender controls after cross-sex hormone treatment.
Collapse
Affiliation(s)
- Lisa A Kilpatrick
- Center for Neurobiology of Stress and Resilience, Department of Medicine, Division of Digestive Diseases, University of California, Los Angeles, California, USA
| | - Mats Holmberg
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden.,ANOVA, Center of Expertise in Andrology, Sexual Medicine, and Transgender Medicine, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine/Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Amirhosein Manzouri
- Department of Women's and Children's Health, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Ivanka Savic
- Department of Women's and Children's Health, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Department of Neurology, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
35
|
Flores‐Vivaldo YM, Camacho‐Abrego I, Picazo O, Flores G. Pregnancies alters spine number in cortical and subcortical limbic brain regions of old rats. Synapse 2019; 73:e22100. [DOI: 10.1002/syn.22100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Yaredit Margarita Flores‐Vivaldo
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología Benemérita Universidad Autónoma de Puebla Puebla Mexico
- Escuela Superior de Medicina, Instituto Politécnico Nacional Mexico City Mexico
| | - Israel Camacho‐Abrego
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología Benemérita Universidad Autónoma de Puebla Puebla Mexico
| | - Ofir Picazo
- Escuela Superior de Medicina, Instituto Politécnico Nacional Mexico City Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología Benemérita Universidad Autónoma de Puebla Puebla Mexico
| |
Collapse
|
36
|
Estrogen-Dependent Functional Spine Dynamics in Neocortical Pyramidal Neurons of the Mouse. J Neurosci 2019; 39:4874-4888. [PMID: 30992373 DOI: 10.1523/jneurosci.2772-18.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 11/21/2022] Open
Abstract
Surgical ovariectomy has been shown to reduce spine density in hippocampal CA1 pyramidal cells of rodents, and this reduction is reversed by 17β-estradiol (E2) treatment in a model of human estrogen replacement therapy. Here, we report reduction of spine density in apical dendrites of layer 5 pyramidal neurons of several neocortical regions that is reversed by subsequent E2 treatment in ovariectomized (OVX) female Thy1M-EGFP mice. We also found that OVX-associated reduction of spine density in somatosensory cortex was accompanied by a reduction in miniature EPSC (mEPSC) frequency (but not mIPSC frequency), indicating a change in functional synapses. OVX-associated spine loss in somatosensory cortex was also rescued by an agonist of the G-protein-linked estrogen receptor (GPER) but not by agonists of the classic estrogen receptors ERα/ERβ, whereas the opposite selectivity was found in area CA1. Acute treatment of neocortical slices with E2 also rescued the OVX-associated reduction in mEPSC frequency, which could be mimicked by a GPER agonist and abolished by a GPER antagonist. Time-lapse in vivo two-photon imaging showed that OVX-associated reduction in spine density is achieved by both an increase in spine loss rate and a decrease in spine gain rate and that subsequent rescue by E2 reversed both of these processes. Crucially, the spines added after E2 rescue were no more likely to reappear at or nearby the sites of pre-OVX spines than those in control mice treated with vehicle. Thus, a model of estrogen replacement therapy, although restoring spine density and dynamics, does not entirely restore functional connectivity.SIGNIFICANCE STATEMENT Estrogen replacement therapy following menopause or surgical removal of the ovaries is a widespread medical practice, yet little is known about the consequences of such treatment for cells in the brain. Here, we show that estrogen replacement reverses some of the effects of surgical removal of the ovaries on the structure and function of brain cells in the mouse. Yet, importantly, the fine wiring of the brain is not returned to the presurgery state by estrogen treatment, suggesting lasting functional consequences.
Collapse
|
37
|
Sheppard PAS, Choleris E, Galea LAM. Structural plasticity of the hippocampus in response to estrogens in female rodents. Mol Brain 2019; 12:22. [PMID: 30885239 PMCID: PMC6423800 DOI: 10.1186/s13041-019-0442-7] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/11/2019] [Indexed: 01/05/2023] Open
Abstract
It is well established that estrogens affect neuroplasticity in a number of brain regions. In particular, estrogens modulate and mediate spine and synapse formation as well as neurogenesis in the hippocampal formation. In this review, we discuss current research exploring the effects of estrogens on dendritic spine plasticity and neurogenesis with a focus on the modulating factors of sex, age, and pregnancy. Hormone levels, including those of estrogens, fluctuate widely across the lifespan from early life to puberty, through adulthood and into old age, as well as with pregnancy and parturition. Dendritic spine formation and modulation are altered both by rapid (likely non-genomic) and classical (genomic) actions of estrogens and have been suggested to play a role in the effects of estrogens on learning and memory. Neurogenesis in the hippocampus is influenced by age, the estrous cycle, pregnancy, and parity in female rodents. Furthermore, sex differences exist in hippocampal cellular and molecular responses to estrogens and are briefly discussed throughout. Understanding how structural plasticity in the hippocampus is affected by estrogens and how these effects can influence function and be influenced by other factors, such as experience and sex, is critical and can inform future treatments in conditions involving the hippocampus.
Collapse
Affiliation(s)
- Paul A. S. Sheppard
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Elena Choleris
- Department of Psychology & Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1 Canada
| | - Liisa A. M. Galea
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| |
Collapse
|
38
|
Clemens AM, Lenschow C, Beed P, Li L, Sammons R, Naumann RK, Wang H, Schmitz D, Brecht M. Estrus-Cycle Regulation of Cortical Inhibition. Curr Biol 2019; 29:605-615.e6. [PMID: 30744972 DOI: 10.1016/j.cub.2019.01.045] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 12/20/2018] [Accepted: 01/15/2019] [Indexed: 01/03/2023]
Abstract
Female mammals experience cyclical changes in sexual receptivity known as the estrus cycle. Little is known about how estrus affects the cortex, although alterations in sensation, cognition and the cyclical occurrence of epilepsy suggest brain-wide processing changes. We performed in vivo juxtacellular and whole-cell recordings in somatosensory cortex of female rats and found that the estrus cycle potently altered cortical inhibition. Fast-spiking interneurons were strongly activated with social facial touch and varied their ongoing activity with the estrus cycle and estradiol in ovariectomized females, while regular-spiking excitatory neurons did not change. In situ hybridization for estrogen receptor β (Esr2) showed co-localization with parvalbumin-positive (PV+) interneurons in deep cortical layers, mirroring the laminar distribution of our physiological findings. The fraction of neurons positive for estrogen receptor β (Esr2) and PV co-localization (Esr2+PV+) in cortical layer V was increased in proestrus. In vivo and in vitro experiments confirmed that estrogen acts locally to increase fast-spiking interneuron excitability through an estrogen-receptor-β-dependent mechanism.
Collapse
Affiliation(s)
- Ann M Clemens
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, Philippstraße 13, Haus 6, 10115 Berlin, Germany
| | - Constanze Lenschow
- Champalimaud Center for the Unknown, Neurosciences, Avenida Brasília, 1400-038 Lisbon, Portugal
| | - Prateep Beed
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Neuroscience Research Center, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Lanxiang Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Xueyuan Boulevard, 518055 Shenzhen, China
| | - Rosanna Sammons
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Neuroscience Research Center, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Robert K Naumann
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Xueyuan Boulevard, 518055 Shenzhen, China
| | - Hong Wang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Xueyuan Boulevard, 518055 Shenzhen, China
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Neuroscience Research Center, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Brecht
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, Philippstraße 13, Haus 6, 10115 Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Neuroscience Research Center, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
39
|
Alexander BH, Barnes HM, Trimmer E, Davidson AM, Ogola BO, Lindsey SH, Mostany R. Stable Density and Dynamics of Dendritic Spines of Cortical Neurons Across the Estrous Cycle While Expressing Differential Levels of Sensory-Evoked Plasticity. Front Mol Neurosci 2018; 11:83. [PMID: 29615867 PMCID: PMC5864847 DOI: 10.3389/fnmol.2018.00083] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/01/2018] [Indexed: 12/11/2022] Open
Abstract
Periodic oscillations of gonadal hormone levels during the estrous cycle exert effects on the female brain, impacting cognition and behavior. While previous research suggests that changes in hormone levels across the cycle affect dendritic spine dynamics in the hippocampus, little is known about the effects on cortical dendritic spines and previous studies showed contradictory results. In this in vivo imaging study, we investigated the impact of the estrous cycle on the density and dynamics of dendritic spines of pyramidal neurons in the primary somatosensory cortex of mice. We also examined if the induction of synaptic plasticity during proestrus, estrus, and metestrus/diestrus had differential effects on the degree of remodeling of synapses in this brain area. We used chronic two-photon excitation (2PE) microscopy during steady-state conditions and after evoking synaptic plasticity by whisker stimulation at the different stages of the cycle. We imaged apical dendritic tufts of layer 5 pyramidal neurons of naturally cycling virgin young female mice. Spine density, turnover rate (TOR), survival fraction, morphology, and volume of mushroom spines remained unaltered across the estrous cycle, and the values of these parameters were comparable with those of young male mice. However, while whisker stimulation of female mice during proestrus and estrus resulted in increases in the TOR of spines (74.2 ± 14.9% and 75.1 ± 12.7% vs. baseline, respectively), sensory-evoked plasticity was significantly lower during metestrus/diestrus (32.3 ± 12.8%). In males, whisker stimulation produced 46.5 ± 20% increase in TOR compared with baseline—not significantly different from female mice at any stage of the cycle. These results indicate that, while steady-state density and dynamics of dendritic spines of layer 5 pyramidal neurons in the primary somatosensory cortex of female mice are constant during the estrous cycle, the susceptibility of these neurons to sensory-evoked structural plasticity may be dependent on the stage of the cycle. Since dendritic spines are more plastic during proestrus and estrus than during metestrus/diestrus, certain stages of the cycle could be more suitable for forms of memory requiring de novo formation and elimination of spines and other stages for forms of memory where retention and/or repurposing of already existing synaptic connections is more pertinent.
Collapse
Affiliation(s)
- Bailin H Alexander
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Heather M Barnes
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Neuroscience Program, Brain Institute, Tulane University, New Orleans, LA, United States
| | - Emma Trimmer
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Andrew M Davidson
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Brain Institute, Tulane University, New Orleans, LA, United States
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Brain Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
40
|
Wang S, Zhu J, Xu T. 17β-estradiol (E2) promotes growth and stability of new dendritic spines via estrogen receptor β pathway in intact mouse cortex. Brain Res Bull 2017; 137:241-248. [PMID: 29288734 DOI: 10.1016/j.brainresbull.2017.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 12/01/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022]
Abstract
The steroid hormone 17β-estradiol (E2) remodels neural circuits at the synaptic level in the mammalian hippocampus and cortex. However, the underlying mechanism of synapse dynamics remains unclear. To elucidate the mechanism, we traced individual dendritic spines on layer V pyramidal neurons of the primary sensory cortex in adult female mice under E2 intervention using two-photon in vivo imaging microscopy. We confirmed the increase of the spine density upon E2 treatment in the intact mouse cortex. Furthermore, we found that this increase is due to the promotion of spine formation and the stability of newly formed spines. E2 treatment doesn't alter the elimination rate of pre-existing spines. Our results also indicate that the activation of the estrogen receptor β (ERβ) mimics the effects of E2 administration on spine dynamics. Taken together, our findings suggest that estrogen promotes growth and stability of new dendritic spines via the ERβ pathway in the intact cortex of female mice.
Collapse
Affiliation(s)
- Shaofang Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jun Zhu
- Chengdu Military General Hospital, Chengdu, China
| | - Tonghui Xu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China.
| |
Collapse
|
41
|
Hwang DS, Kim N, Choi JG, Kim HG, Kim H, Oh MS. Dangguijakyak-san ameliorates memory deficits in ovariectomized mice by upregulating hippocampal estrogen synthesis. Altern Ther Health Med 2017; 17:501. [PMID: 29178947 PMCID: PMC5702078 DOI: 10.1186/s12906-017-2015-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 11/17/2017] [Indexed: 01/20/2023]
Abstract
Background Dangguijakyak-san (DJS) is an herbal formulation that has been clinically applicable for treating postmenopausal symptoms and neurological disorders. It is reported that hippocampal estrogen attenuates memory impairment via neuroprotection and synaptogenesis. However, the effect of DJS on hippocampal estrogen synthesis remains unknown. In this study, we explored the effect of DJS and its neuroprotective mechanism against memory impairment in ovariectomized (OVX) mice, with respect to hippocampal estrogen stimulation. Methods Cell cultures were prepared from the hippocampi of 18-day-old embryos from timed pregnant Sprague–Dawley rats. The hippocampi were dissected, collected, dissociated, and plated in 60-mm dishes. The cells were treated with DJS for 48 h and the supernatant was collected to determine estrogen levels. Female ICR mice (8-weeks-old) were housed for 1 week and ovariectomy was performed to remove the influence of ovary-synthesized estrogens. Following a 2-week post-surgical recovery period, the mice were administrated with DJS (50 and 100 mg/kg/day, p.o.) or 17β-estradiol (200 μg/kg/day, i.p.) once daily for 21 days. Hippocampal and serum estrogen levels were determined using enzyme-linked immunosorbent assay kit. Memory behavioral tests, western blot, and immunohistochemical analyses were performed to evaluate the neuroprotective effects of DJS in this model. Results DJS treatment promoted estrogen synthesis in primary hippocampal cells and the hippocampus of OVX mice, resulting in the amelioration of OVX-induced memory impairment. Hippocampal estrogen stimulated by DJS treatment contributed to the activation of cAMP response element-binding protein and synaptic protein in OVX mice. Conclusion DJS may attenuate memory deficits in postmenopausal women via hippocampal estrogen synthesis. Electronic supplementary material The online version of this article (10.1186/s12906-017-2015-6) contains supplementary material, which is available to authorized users.
Collapse
|
42
|
The Progestin Receptor Interactome in the Female Mouse Hypothalamus: Interactions with Synaptic Proteins Are Isoform Specific and Ligand Dependent. eNeuro 2017; 4:eN-NWR-0272-17. [PMID: 28955722 PMCID: PMC5605756 DOI: 10.1523/eneuro.0272-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/31/2017] [Accepted: 09/01/2017] [Indexed: 01/06/2023] Open
Abstract
Progestins bind to the progestin receptor (PR) isoforms, PR-A and PR-B, in brain to influence development, female reproduction, anxiety, and stress. Hormone-activated PRs associate with multiple proteins to form functional complexes. In the present study, proteins from female mouse hypothalamus that associate with PR were isolated using affinity pull-down assays with glutathione S-transferase–tagged mouse PR-A and PR-B. Using complementary proteomics approaches, reverse phase protein array (RPPA) and mass spectrometry, we identified hypothalamic proteins that interact with PR in a ligand-dependent and isoform-specific manner and were confirmed by Western blot. Synaptic proteins, including synapsin-I and synapsin-II, interacted with agonist-bound PR isoforms, suggesting that both isoforms function in synaptic plasticity. In further support, synaptogyrin-III and synapsin-III associated with PR-A and PR-B, respectively. PR also interacted with kinases, including c-Src, mTOR, and MAPK1, confirming phosphorylation as an integral process in rapid effects of PR in the brain. Consistent with a role in transcriptional regulation, PR associated with transcription factors and coactivators in a ligand-specific and isoform-dependent manner. Interestingly, both PR isoforms associated with a key regulator of energy homeostasis, FoxO1, suggesting a novel role for PR in energy metabolism. Because many identified proteins in this PR interactome are synaptic proteins, we tested the hypothesis that progestins function in synaptic plasticity. Indeed, progesterone enhanced synaptic density, by increasing synapsin-I–positive synapses, in rat primary cortical neuronal cultures. This novel combination of RPPA and mass spectrometry allowed identification of PR action in synaptic remodeling and energy homeostasis and reveals unique roles for progestins in brain function and disease.
Collapse
|
43
|
Abstract
Contrary to popular belief, sex hormones act throughout the entire brain of both males and females via both genomic and nongenomic receptors. Many neural and behavioral functions are affected by estrogens, including mood, cognitive function, blood pressure regulation, motor coordination, pain, and opioid sensitivity. Subtle sex differences exist for many of these functions that are developmentally programmed by hormones and by not yet precisely defined genetic factors, including the mitochondrial genome. These sex differences, and responses to sex hormones in brain regions and upon functions not previously regarded as subject to such differences, indicate that we are entering a new era in our ability to understand and appreciate the diversity of gender-related behaviors and brain functions.
Collapse
Affiliation(s)
- Jordan Marrocco
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York, USA
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York, USA
| |
Collapse
|
44
|
McEwen BS, Milner TA. Understanding the broad influence of sex hormones and sex differences in the brain. J Neurosci Res 2017; 95:24-39. [PMID: 27870427 PMCID: PMC5120618 DOI: 10.1002/jnr.23809] [Citation(s) in RCA: 356] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/23/2016] [Accepted: 06/10/2016] [Indexed: 12/11/2022]
Abstract
Sex hormones act throughout the entire brain of both males and females via both genomic and nongenomic receptors. Sex hormones can act through many cellular and molecular processes that alter structure and function of neural systems and influence behavior as well as providing neuroprotection. Within neurons, sex hormone receptors are found in nuclei and are also located near membranes, where they are associated with presynaptic terminals, mitochondria, spine apparatus, and postsynaptic densities. Sex hormone receptors also are found in glial cells. Hormonal regulation of a variety of signaling pathways as well as direct and indirect effects on gene expression induce spine synapses, up- or downregulate and alter the distribution of neurotransmitter receptors, and regulate neuropeptide expression and cholinergic and GABAergic activity as well as calcium sequestration and oxidative stress. Many neural and behavioral functions are affected, including mood, cognitive function, blood pressure regulation, motor coordination, pain, and opioid sensitivity. Subtle sex differences exist for many of these functions that are developmentally programmed by hormones and by not yet precisely defined genetic factors, including the mitochondrial genome. These sex differences and responses to sex hormones in brain regions, which influence functions not previously regarded as subject to such differences, indicate that we are entering a new era of our ability to understand and appreciate the diversity of gender-related behaviors and brain functions. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bruce S. McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Teresa A. Milner
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
- Feil Family Brain and Mind Research Institute, Weill Cornell School of Medicine, 407 East 61st Street, New York, NY 10065
| |
Collapse
|
45
|
Chen JR, Lim SH, Chung SC, Lee YF, Wang YJ, Tseng GF, Wang TJ. Reproductive experience modified dendritic spines on cortical pyramidal neurons to enhance sensory perception and spatial learning in rats. Exp Anim 2016; 66:61-74. [PMID: 27784858 PMCID: PMC5301002 DOI: 10.1538/expanim.16-0061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Behavioral adaptations during motherhood are aimed at increasing reproductive success.
Alterations of hormones during motherhood could trigger brain morphological changes to
underlie behavioral alterations. Here we investigated whether motherhood changes a rat’s
sensory perception and spatial memory in conjunction with cortical neuronal structural
changes. Female rats of different statuses, including virgin, pregnant, lactating, and
primiparous rats were studied. Behavioral test showed that the lactating rats were most
sensitive to heat, while rats with motherhood and reproduction experience outperformed
virgin rats in a water maze task. By intracellular dye injection and computer-assisted
3-dimensional reconstruction, the dendritic arbors and spines of the layer III and V
pyramidal neurons of the somatosensory cortex and CA1 hippocampal pyramidal neurons were
revealed for closer analysis. The results showed that motherhood and reproductive
experience increased dendritic spines but not arbors or the lengths of the layer III and V
pyramidal neurons of the somatosensory cortex and CA1 hippocampal pyramidal neurons. In
addition, lactating rats had a higher incidence of spines than pregnant or primiparous
rats. The increase of dendritic spines was coupled with increased expression of the
glutamatergic postsynaptic marker protein (PSD-95), especially in lactating rats. On the
basis of the present results, it is concluded that motherhood enhanced rat sensory
perception and spatial memory and was accompanied by increases in dendritic spines on
output neurons of the somatosensory cortex and CA1 hippocampus. The effect was sustained
for at least 6 weeks after the weaning of the pups.
Collapse
Affiliation(s)
- Jeng-Rung Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
46
|
White EC, Graham BM. Estradiol levels in women predict skin conductance response but not valence and expectancy ratings in conditioned fear extinction. Neurobiol Learn Mem 2016; 134 Pt B:339-48. [DOI: 10.1016/j.nlm.2016.08.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 07/31/2016] [Accepted: 08/17/2016] [Indexed: 01/03/2023]
|
47
|
Sarkar A, Kabbaj M. Sex Differences in Effects of Ketamine on Behavior, Spine Density, and Synaptic Proteins in Socially Isolated Rats. Biol Psychiatry 2016; 80:448-456. [PMID: 26957131 PMCID: PMC4940294 DOI: 10.1016/j.biopsych.2015.12.025] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/18/2015] [Accepted: 12/30/2015] [Indexed: 01/12/2023]
Abstract
BACKGROUND The mechanistic underpinnings of sex differences in occurrence of depression and efficacy of antidepressant treatments are poorly understood. We examined the effects of isolation stress (IS) and the fast-acting antidepressant ketamine on anhedonia and depression-like behavior, spine density, and synaptic proteins in male and female rats. METHODS We used a chronic social IS paradigm to test the effects of ketamine (0, 2.5 mg/kg, and 5 mg/kg) on behavior and levels of synaptic proteins synapsin-1, postsynaptic density protein 95, and glutamate receptor 1 in male rats and female rats in diestrus. Medial prefrontal cortex spine density was also examined in male rats and female rats that received ketamine during either the diestrus or the proestrus phase of their estrous cycle. RESULTS Male rats showed anhedonia and depression-like behavior after 8 weeks of IS, concomitant with decreases in spine density and levels of synapsin-1, postsynaptic density protein 95, and glutamate receptor 1 in the medial prefrontal cortex; these changes were reversed by a single injection of ketamine (5 mg/kg). After 11 weeks of IS, female rats showed depression-like behavior but no signs of anhedonia. Although both doses of ketamine rescued depression-like behavior in female rats, the decline observed in synaptic proteins and spine density in IS and in diestrus female rats could not be reversed by ketamine. Spine density was higher in female rats during proestrus than in diestrus. CONCLUSIONS Our findings implicate a role for synaptic proteins synapsin-1, postsynaptic density protein 95, and glutamate receptor 1 and medial prefrontal cortex spine density in the antidepressant effects of ketamine in male rats subjected to IS but not in female rats subjected to IS, suggesting dissimilar underlying mechanisms for efficacy of ketamine in the two sexes.
Collapse
Affiliation(s)
- Ambalika Sarkar
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Program in Neuroscience, College of Medicine, Florida State University, Tallahassee, Florida.
| |
Collapse
|
48
|
Chen LJ, Wang YJ, Chen JR, Tseng GF. Hydrocephalus compacted cortex and hippocampus and altered their output neurons in association with spatial learning and memory deficits in rats. Brain Pathol 2016; 27:419-436. [PMID: 27411167 DOI: 10.1111/bpa.12414] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/04/2016] [Indexed: 12/29/2022] Open
Abstract
Hydrocephalus is a common neurological disorder in children characterized by abnormal dilation of cerebral ventricles as a result of the impairment of cerebrospinal fluid flow or absorption. Clinical presentation of hydrocephalus varies with chronicity and often shows cognitive dysfunction. Here we used a kaolin-induction method in rats and studied the effects of hydrocephalus on cerebral cortex and hippocampus, the two regions highly related to cognition. Hydrocephalus impaired rats' performance in Morris water maze task. Serial three-dimensional reconstruction from sections of the whole brain freshly froze in situ with skull shows that the volumes of both structures were reduced. Morphologically, pyramidal neurons of the somatosensory cortex and hippocampus appear to be distorted. Intracellular dye injection and subsequent three-dimensional reconstruction and analyses revealed that the dendritic arbors of layer III and V cortical pyramid neurons were reduced. The total dendritic length of CA1, but not CA3, pyramidal neurons was also reduced. Dendritic spine densities on both cortical and hippocampal pyramidal neurons were decreased, consistent with our concomitant findings that the expressions of both synaptophysin and postsynaptic density protein 95 were reduced. These cortical and hippocampal changes suggest reductions of excitatory connectivity, which could underlie the learning and memory deficits in hydrocephalus.
Collapse
Affiliation(s)
- Li-Jin Chen
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien City, Taiwan
| | - Yueh-Jan Wang
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien City, Taiwan
| | - Jeng-Rung Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Guo-Fang Tseng
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien City, Taiwan
| |
Collapse
|
49
|
Vigil P, Del Río JP, Carrera BÁ, ArÁnguiz FC, Rioseco H, Cortés ME. Influence of sex steroid hormones on the adolescent brain and behavior: An update. LINACRE QUARTERLY 2016; 83:308-329. [PMID: 27833209 DOI: 10.1080/00243639.2016.1211863] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This review explains the main effects exerted by sex steroids and other hormones on the adolescent brain. During the transition from puberty to adolescence, these hormones participate in the organizational phenomena that structurally shape some brain circuits. In adulthood, this will propitiate some specific behavior as responses to the hormones now activating those neural circuits. Adolescence is, then, a critical "organizational window" for the brain to develop adequately, since steroid hormones perform important functions at this stage. For this reason, the adolescent years are very important for future behaviors in human beings. Changes that occur or fail to occur during adolescence will determine behaviors for the rest of one's lifetime. Consequently, understanding the link between adolescent behavior and brain development as influenced by sex steroids and other hormones and compounds is very important in order to interpret various psycho-affective pathologies. Lay Summary : The effect of steroid hormones on the development of the adolescent brain, and therefore, on adolescent behavior, is noticeable. This review presents their main activational and organizational effects. During the transition from puberty to adolescence, organizational phenomena triggered by steroids structurally affect the remodeling of brain circuits. Later in adulthood, these changes will be reflected in behavioral responses to such hormones. Adolescence can then be seen as a fundamental "organizational window" during which sex steroids and other hormones and compounds play relevant roles. The understanding of the relationship between adolescent behavior and the way hormones influence brain development help understand some psychological disorders.
Collapse
Affiliation(s)
- Pilar Vigil
- Pontificia Universidad Católica de Chile, Vicerrectoría de Comunicaciones, Santiago, Chile; Biomedical Division, Reproductive Health Research Institute, Santiago, Chile
| | - Juan Pablo Del Río
- Biomedical Division, Reproductive Health Research Institute, Santiago, Chile; Universidad de los Andes, Facultad de Medicina, Escuela de Medicina, Santiago, Chile
| | - BÁrbara Carrera
- Biomedical Division, Reproductive Health Research Institute, Santiago, Chile
| | | | - Hernán Rioseco
- Biomedical Division, Reproductive Health Research Institute, Santiago, Chile
| | - Manuel E Cortés
- Biomedical Division, Reproductive Health Research Institute, Santiago, Chile; Universidad Bernardo O Higgins, Facultad de Salud, Departamento de Ciencias Químicas y Biológicas, Santiago, Chile
| |
Collapse
|
50
|
Ferreira R, Brandão ML, Nobre MJ. 5-HT1A receptors of the prelimbic cortex mediate the hormonal impact on learned fear expression in high-anxious female rats. Horm Behav 2016; 84:84-96. [PMID: 27328163 DOI: 10.1016/j.yhbeh.2016.05.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 05/13/2016] [Accepted: 05/19/2016] [Indexed: 12/26/2022]
Abstract
Hormones highly influence female behaviors. However, research on this topic has not usually considered the variable hormonal status. The prelimbic cortex (PrL) is commonly engaged in fear learning. Connections from and to this region are known to be critical in regulating anxiety, in which serotonin (5-HT) plays a fundamental role, particularly through changes in 5-HT1A receptors functioning. Also, hormone fluctuations can greatly influence anxiety in humans and anxiety-related behavior in rodents, and this influence involves the functioning of 5-HT brain systems. The present investigation sought to determine whether fluctuations in ovarian hormones relative to the estrous cycle would influence the expression of learned fear in female rats previously selected as low- (LA) or high-anxious (HA). Furthermore, we investigate the role of the 5-HT system of the PrL, particularly the 5-HT1A receptors, as a possible modulator of estrous cycle influence on the expression of learned fear through intra-PrL microinjections of 5-HT itself or the full 5-HT1A agonist 8-OH-DPAT (8-hydroxy-2-(di-n-propylamine)tetralin). Behavioral changes were assessed using the fear-potentiated startle (FPS) procedure. The results showed that fear intensity is associated with hormonal decay, being more accentuated during the estrus phase. This increase in fear levels was found to be negatively correlated with the expression of potentiated startle. In rats prone to anxiety and tested during the proestrus and estrus phases, 5-HT mechanisms of the PrL seem to play a regulatory role in the expression of learned fear. These results were not replicated in the LA rats. Similar but less intense results were found regarding the early and late diestrus. Our data indicate that future studies on this subject need to take into account the dissociation between low- and high-responsive females to understand how hormones affect emotional behavior.
Collapse
Affiliation(s)
- Renata Ferreira
- Departamento de Psicologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), 14040-901 Ribeirão Preto, SP, Brazil; Instituto de Neurociências e Comportamento-INeC, Campus USP, 14040-901 Ribeirão Preto, SP, Brazil
| | - Marcus Lira Brandão
- Departamento de Psicologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), 14040-901 Ribeirão Preto, SP, Brazil; Instituto de Neurociências e Comportamento-INeC, Campus USP, 14040-901 Ribeirão Preto, SP, Brazil
| | - Manoel Jorge Nobre
- Departamento de Psicologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), 14040-901 Ribeirão Preto, SP, Brazil; Departamento de Psicologia, Uni-FACEF, 14401-135 Franca, SP, Brazil; Instituto de Neurociências e Comportamento-INeC, Campus USP, 14040-901 Ribeirão Preto, SP, Brazil.
| |
Collapse
|