1
|
Livia R, Kim H, Emily M, Luise MM, Haiko S, Julia S. "Estrogens and human brain networks: A systematic review of structural and functional neuroimaging studies". Front Neuroendocrinol 2024:101174. [PMID: 39733923 DOI: 10.1016/j.yfrne.2024.101174] [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: 06/17/2024] [Revised: 10/23/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024]
Abstract
Estrogen fluctuations during the menstrual cycle, puberty, postpartum, or in the menopausal transition are associated with cognitive, affective, and behavioral effects. Additionally, estrogens are essential in hormonal contraception, menopausal hormone therapy, or gender-affirming hormone therapy. This systematic review summarizes findings on the role of estrogens for structure, function, and connectivity of human brain networks. We searched PubMed, Web of Science, and ScienceDirect for neuroimaging articles assessing estrogens published since 2008. We included 54 studies (N = 2,494 participants) on endogenous estrogen, and 28 studies (N = 1740 participants) on exogenous estrogen conditions. Estrogen-related changes were reported for emotion, reward, memory, and resting-state networks, and in regional white and gray matter, with a particular neural plasticity in the hippocampus and amygdala. By examining study designs, imaging measures, and analysis methods, this review highlights the role of neuroimaging in advancing neuroendocrine and neurocognitive research, particularly promoting brain health for women and individuals with ovaries.
Collapse
Affiliation(s)
- Ruehr Livia
- Centre for Integrative Women's Health and Gender Medicine, Medical Faculty & University Hospital Leipzig, Germany; Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1A, 04103 Leipzig, Germany; Max Planck School of Cognition, Stephanstraße 1A, 04103 Leipzig, Germany; Cognitive Neurology, University Medical Center Leipzig, Liebigstraße 16, 04103 Leipzig, Germany.
| | - Hoffmann Kim
- Centre for Integrative Women's Health and Gender Medicine, Medical Faculty & University Hospital Leipzig, Germany; Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1A, 04103 Leipzig, Germany; Cognitive Neurology, University Medical Center Leipzig, Liebigstraße 16, 04103 Leipzig, Germany; Humboldt-Universität zu Berlin, Berlin School of Mind and Brain, Unter den Linden 6, 10099 Berlin, Germany.
| | - May Emily
- Centre for Integrative Women's Health and Gender Medicine, Medical Faculty & University Hospital Leipzig, Germany; Max Planck School of Cognition, Stephanstraße 1A, 04103 Leipzig, Germany; Cognitive Neurology, University Medical Center Leipzig, Liebigstraße 16, 04103 Leipzig, Germany.
| | - Münch Marie Luise
- Leipzig Reproductive Health Research Center, Liebigstraße 20A, 04103 Leipzig, Germany.
| | - Schlögl Haiko
- Department of Endocrinology, Nephrology, Rheumatology, Division of Endocrinology, University Hospital Leipzig, Liebigstraße 20, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Straße 27, 04103 Leipzig, Germany.
| | - Sacher Julia
- Centre for Integrative Women's Health and Gender Medicine, Medical Faculty & University Hospital Leipzig, Germany; Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1A, 04103 Leipzig, Germany; Max Planck School of Cognition, Stephanstraße 1A, 04103 Leipzig, Germany; Cognitive Neurology, University Medical Center Leipzig, Liebigstraße 16, 04103 Leipzig, Germany; Department of Endocrinology, Nephrology, Rheumatology, Division of Endocrinology, University Hospital Leipzig, Liebigstraße 20, 04103 Leipzig, Germany.
| |
Collapse
|
2
|
Kapri D, Pradhan A, Vuruputuri RM, Vaidya VA. Sex differences in the influence of adult-onset hypothyroidism on hippocampal progenitor survival and neuronal differentiation in mice. J Neuroendocrinol 2024; 36:e13453. [PMID: 39360641 DOI: 10.1111/jne.13453] [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: 11/27/2023] [Revised: 08/29/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
The ongoing production of newborn neurons in the adult hippocampus is reported to be sensitive to perturbations of thyroid hormone signaling, in male rats and mice. Here, we examined whether the neurogenic changes evoked by adult-onset hypothyroidism exhibit sex differences, using male and female C57BL/6N mice. We assessed the impact of goitrogen-induced, adult-onset hypothyroidism on the postmitotic survival and differentiation of hippocampal progenitors in male and female mice. Adult-onset hypothyroidism evoked a significant decline in the postmitotic survival and neuronal differentiation of adult-born progenitors within the dentate gyrus hippocampal subfield of male, but not female, mice. We observed a significant decrease in the number of immature neurons within the hippocampi of adult-onset hypothyroid male mice, whereas adult-onset hypothyroidism evoked by goitrogens using the same treatment paradigms did not evoke any change in immature neuron number in female mice. Gene expression analysis within the hippocampi of euthyroid male and female mice revealed sex-dependent, differential expression of thyroid hormone receptor genes, as well as genes linked to thyroid hormone metabolism and transport. Collectively, our findings highlight sex differences in the influence of goitrogen-induced, adult-onset hypothyroidism on hippocampal neurogenesis, with male, but not female, mice exhibiting a decline in postmitotic hippocampal progenitor survival and neuronal differentiation. These findings underscore the importance of sex as a vital variable when considering the impact of thyroid hormone signaling on the adult hippocampal neurogenic niche.
Collapse
Affiliation(s)
- Darshana Kapri
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India
| | - Amartya Pradhan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India
| | - Ratna Mahathi Vuruputuri
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra, India
| |
Collapse
|
3
|
Martínez-Martos JM, Cantón-Habas V, Rich-Ruíz M, Reyes-Medina MJ, Ramírez-Expósito MJ, Carrera-González MDP. Sexual and Metabolic Differences in Hippocampal Evolution: Alzheimer's Disease Implications. Life (Basel) 2024; 14:1547. [PMID: 39768255 PMCID: PMC11677427 DOI: 10.3390/life14121547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Sex differences in brain metabolism and their relationship to neurodegenerative diseases like Alzheimer's are an important emerging topic in neuroscience. Intrinsic anatomic and metabolic differences related to male and female physiology have been described, underscoring the importance of considering biological sex in studying brain metabolism and associated pathologies. The hippocampus is a key structure exhibiting sex differences in volume and connectivity. Adult neurogenesis in the dentate gyrus, dendritic spine density, and electrophysiological plasticity contribute to the hippocampus' remarkable plasticity. Glucose transporters GLUT3 and GLUT4 are expressed in human hippocampal neurons, with proper glucose metabolism being crucial for learning and memory. Sex hormones play a major role, with the aromatase enzyme that generates estradiol increasing in neurons and astrocytes as an endogenous neuroprotective mechanism. Inhibition of aromatase increases gliosis and neurodegeneration after brain injury. Genetic variants of aromatase may confer higher Alzheimer's risk. Estrogen replacement therapy in postmenopausal women prevents hippocampal hypometabolism and preserves memory. Insulin is also a key regulator of hippocampal glucose metabolism and cognitive processes. Dysregulation of the insulin-sensitive glucose transporter GLUT4 may explain the comorbidity between type II diabetes and Alzheimer's. GLUT4 colocalizes with the insulin-regulated aminopeptidase IRAP in neuronal vesicles, suggesting an activity-dependent glucose uptake mechanism. Sex differences in brain metabolism are an important factor in understanding neurodegenerative diseases, and future research must elucidate the underlying mechanisms and potential therapeutic implications of these differences.
Collapse
Affiliation(s)
- José Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Las Lagunillas University Campus, 23009 Jaen, Spain; (J.M.M.-M.); (M.J.R.-E.)
| | - Vanesa Cantón-Habas
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, University of Córdoba, 14004 Córdoba, Spain; (V.C.-H.); (M.R.-R.); (M.J.R.-M.)
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14004 Cordoba, Spain
| | - Manuel Rich-Ruíz
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, University of Córdoba, 14004 Córdoba, Spain; (V.C.-H.); (M.R.-R.); (M.J.R.-M.)
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14004 Cordoba, Spain
| | - María José Reyes-Medina
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, University of Córdoba, 14004 Córdoba, Spain; (V.C.-H.); (M.R.-R.); (M.J.R.-M.)
| | - María Jesús Ramírez-Expósito
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Las Lagunillas University Campus, 23009 Jaen, Spain; (J.M.M.-M.); (M.J.R.-E.)
| | - María del Pilar Carrera-González
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Las Lagunillas University Campus, 23009 Jaen, Spain; (J.M.M.-M.); (M.J.R.-E.)
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14004 Cordoba, Spain
| |
Collapse
|
4
|
Yagi S, Mohammad A, Wen Y, Batallán Burrowes AA, Blankers SA, Galea LAM. Estrogens dynamically regulate neurogenesis in the dentate gyrus of adult female rats. Hippocampus 2024; 34:583-597. [PMID: 39166359 DOI: 10.1002/hipo.23633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Estrone and estradiol differentially modulate neuroplasticity and cognition. How they influence the maturation of new neurons in the adult hippocampus, however, is not known. The present study assessed the effects of estrone and estradiol on the maturation timeline of neurogenesis in the dentate gyrus (DG) of ovariectomized (a model of surgical menopause) young adult Sprague-Dawley rats using daily subcutaneous injections of 17β-estradiol, estrone or vehicle. Rats were injected with a DNA synthesis marker, 5-bromo-2-deoxyuridine (BrdU), and were perfused 1, 2, or 3 weeks after BrdU injection and daily hormone treatment. Brains were sectioned and processed for various markers including: sex-determining region Y-box 2 (Sox2), glial fibrillary acidic protein (GFAP), antigen kiel 67 (Ki67), doublecortin (DCX), and neuronal nuclei (NeuN). Immunofluorescent labeling or co-labelling of BrdU with Sox2 (progenitor cells), Sox2/GFAP (neural progenitor cells), Ki67 (cell proliferation), DCX (immature neurons), NeuN (mature neurons) was used to examine the trajectory and maturation of adult-born neurons over time. Estrogens had early (1 week of exposure) effects on different stages of neurogenesis (neural progenitor cells, cell proliferation and early maturation of new cells into neurons) but these effects were less pronounced after prolonged treatment. Estradiol enhanced, whereas estrone reduced cell proliferation after 1 week but not after longer exposure to either estrogen. Both estrogens increased the density of immature neurons (BrdU/DCX-ir) after 1 week of exposure compared to vehicle treatment but this increased density was not sustained over longer durations of treatments to estrogens, suggesting that the enhancing effects of estrogens on neurogenesis were short-lived. Longer duration post-ovariectomy, without treatments with either of the estrogens, was associated with reduced neural progenitor cells in the DG. These results demonstrate that estrogens modulate several aspects of adult hippocampal neurogenesis differently in the short term, but may lose their ability to influence neurogenesis after long-term exposure. These findings have potential implications for treatments involving estrogens after surgical menopause.
Collapse
Affiliation(s)
- Shunya Yagi
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ahmad Mohammad
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Yanhua Wen
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ariel A Batallán Burrowes
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Garcia-Segura LM, Méndez P, Arevalo MA, Azcoitia I. Neuroestradiol and neuronal development: Not an exclusive male tale anymore. Front Neuroendocrinol 2023; 71:101102. [PMID: 37689249 DOI: 10.1016/j.yfrne.2023.101102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
The brain synthesizes a variety of neurosteroids, including neuroestradiol. Inhibition of neuroestradiol synthesis results in alterations in basic neurodevelopmental processes, such as neurogenesis, neuroblast migration, neuritogenesis and synaptogenesis. Although the neurodevelopmental actions of neuroestradiol are exerted in both sexes, some of them are sex-specific, such as the well characterized effects of neuroestradiol derived from the metabolism of testicular testosterone during critical periods of male brain development. In addition, recent findings have shown sex-specific actions of neuroestradiol on neuroblast migration, neuritic growth and synaptogenesis in females. Among other factors, the epigenetic regulation exerted by X linked genes, such as Kdm6a/Utx, may determine sex-specific actions of neuroestradiol in the female brain. This review evidences the impact of neuroestradiol on brain formation in both sexes and highlights the interaction of neural steriodogenesis, hormones and sex chromosomes in sex-specific brain development.
Collapse
Affiliation(s)
- Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Nacional de Salud Carlos III, Madrid, Spain.
| | - Pablo Méndez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - M Angeles Arevalo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Nacional de Salud Carlos III, Madrid, Spain.
| | - Iñigo Azcoitia
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Nacional de Salud Carlos III, Madrid, Spain; Department of Cell Biology, Universidad Complutense de Madrid, C José Antonio Nováis 12, 28040 Madrid, Spain
| |
Collapse
|
6
|
Yagi S, Lieblich SE, Galea LAM. High estradiol reduces adult neurogenesis but strengthens functional connectivity within the hippocampus during spatial pattern separation in adult female rats. Horm Behav 2023; 155:105409. [PMID: 37567060 DOI: 10.1016/j.yhbeh.2023.105409] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023]
Abstract
Adult neurogenesis in the dentate gyrus plays an important role for pattern separation, the process of separating similar inputs and forming distinct neural representations. Estradiol modulates neurogenesis and hippocampus function, but to date no examination of estradiol's effects on pattern separation have been conducted. Here, we examined estrogenic regulation of adult neurogenesis and functional connectivity in the hippocampus after the spatial pattern separation task in female rats. Ovariectomized Sprague-Dawley rats received daily injections of vehicle, 0.32 μg (Low) or 5 μg (High) of estradiol benzoate until the end of experiment. A single bromodeoxyuridine (BrdU) was injected one day after initiation of hormone or vehicle treatment and rats were tested in the delayed nonmatching to position spatial pattern separation task in the 8-arm radial maze for 12 days beginning two weeks after BrdU injection. Rats were perfused 90 min after the final trial and brain sections were immunohistochemically stained for BrdU/neuronal nuclei (NeuN) (new neurons), Ki67 (cell proliferation), and the immediate early gene, zif268 (activation). Results showed that high, but not low, estradiol reduced the density of BrdU/NeuN-ir cells and had significant inter-regional correlations of zif268-ir cell density in the hippocampus following pattern separation. Estradiol treatment did not influence pattern separation performance or strategy use. These results show that higher doses of estradiol can reduce neurogenesis but at the same time increases correlations of activity of neurons within the hippocampus during spatial pattern separation.
Collapse
Affiliation(s)
- Shunya Yagi
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | | | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Department of Psychology, University of British Columbia, Vancouver, Canada; Djavad Mowifaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
7
|
Calvo N, Einstein G. Steroid hormones: risk and resilience in women's Alzheimer disease. Front Aging Neurosci 2023; 15:1159435. [PMID: 37396653 PMCID: PMC10313425 DOI: 10.3389/fnagi.2023.1159435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
More women have Alzheimer disease (AD) than men, but the reasons for this phenomenon are still unknown. Including women in clinical research and studying their biology is key to understand not just their increased risk but also their resilience against the disease. In this sense, women are more affected by AD than men, but their reserve or resilience mechanisms might delay symptom onset. The aim of this review was to explore what is known about mechanisms underlying women's risk and resilience in AD and identify emerging themes in this area that merit further research. We conducted a review of studies analyzing molecular mechanisms that may induce neuroplasticity in women, as well as cognitive and brain reserve. We also analyzed how the loss of steroid hormones in aging may be linked to AD. We included empirical studies with human and animal models, literature reviews as well as meta-analyses. Our search identified the importance of 17-b-estradiol (E2) as a mechanism driving cognitive and brain reserve in women. More broadly, our analysis revealed the following emerging perspectives: (1) the importance of steroid hormones and their effects on both neurons and glia for the study of risk and resilience in AD, (2) E2's crucial role in women's brain reserve, (3) women's verbal memory advantage as a cognitive reserve factor, and (4) E2's potential role in linguistic experiences such as multilingualism and hearing loss. Future directions for research include analyzing the reserve mechanisms of steroid hormones on neuronal and glial plasticity, as well as identifying the links between steroid hormone loss in aging and risk for AD.
Collapse
Affiliation(s)
- Noelia Calvo
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Gillian Einstein
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON, Canada
- Tema Genus, Linköping University, Linköping, Sweden
- Women’s College Research Institute, Toronto, ON, Canada
- Centre for Life Course and Aging, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Effects of exogenous oxytocin and estradiol on resting-state functional connectivity in women and men. Sci Rep 2023; 13:3113. [PMID: 36813823 PMCID: PMC9947123 DOI: 10.1038/s41598-023-29754-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Possible interactions of the neuropeptide oxytocin and the sex hormone estradiol may contribute to previously observed sex-specific effects of oxytocin on resting-state functional connectivity (rsFC) of the amygdala and hippocampus. Therefore, we used a placebo-controlled, randomized, parallel-group functional magnetic resonance imaging study design and measured amygdala and hippocampus rsFC in healthy men (n = 116) and free-cycling women (n = 111), who received estradiol gel (2 mg) or placebo before the intranasal administration of oxytocin (24 IU) or placebo. Our results reveal significant interaction effects of sex and treatments on rsFC of the amygdala and hippocampus in a seed-to-voxel analysis. In men, both oxytocin and estradiol significantly decreased rsFC between the left amygdala and the right and left lingual gyrus, the right calcarine fissure, and the right superior parietal gyrus compared to placebo, while the combined treatment produced a significant increase in rsFC. In women, the single treatments significantly increased the rsFC between the right hippocampus and the left anterior cingulate gyrus, whereas the combined treatment had the opposite effect. Collectively, our study indicates that exogenous oxytocin and estradiol have different region-specific effects on rsFC in women and men and that the combined treatment may produce antagonistic effects.
Collapse
|
9
|
Turek J, Gąsior Ł. Estrogen fluctuations during the menopausal transition are a risk factor for depressive disorders. Pharmacol Rep 2023; 75:32-43. [PMID: 36639604 PMCID: PMC9889489 DOI: 10.1007/s43440-022-00444-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023]
Abstract
Women are significantly more likely to develop depression than men. Fluctuations in the ovarian estrogen hormone levels are closely linked with women's well-being. This narrative review discusses the available knowledge on the role of estrogen in modulating brain function and the correlation between changes in estrogen levels and the development of depression. Equally discussed are the possible mechanisms underlying these effects, including the role of estrogen in modulating brain-derived neurotrophic factor activity, serotonin neurotransmission, as well as the induction of inflammatory response and changes in metabolic activity, are discussed.
Collapse
Affiliation(s)
- Justyna Turek
- Department of Neurobiology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland
| | - Łukasz Gąsior
- Department of Neurobiology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland
| |
Collapse
|
10
|
Harrington YA, Parisi JM, Duan D, Rojo-Wissar DM, Holingue C, Spira AP. Sex Hormones, Sleep, and Memory: Interrelationships Across the Adult Female Lifespan. Front Aging Neurosci 2022; 14:800278. [PMID: 35912083 PMCID: PMC9331168 DOI: 10.3389/fnagi.2022.800278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 06/09/2022] [Indexed: 01/26/2023] Open
Abstract
As the population of older adults grows, so will the prevalence of aging-related conditions, including memory impairments and sleep disturbances, both of which are more common among women. Compared to older men, older women are up to twice as likely to experience sleep disturbances and are at a higher risk of cognitive decline and Alzheimer's disease and related dementias (ADRD). These sex differences may be attributed in part to fluctuations in levels of female sex hormones (i.e., estrogen and progesterone) that occur across the adult female lifespan. Though women tend to experience the most significant sleep and memory problems during the peri-menopausal period, changes in memory and sleep have also been observed across the menstrual cycle and during pregnancy. Here, we review current knowledge on the interrelationships among female sex hormones, sleep, and memory across the female lifespan, propose possible mediating and moderating mechanisms linking these variables and describe implications for ADRD risk in later life.
Collapse
Affiliation(s)
- Yasmin A. Harrington
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jeanine M. Parisi
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Daisy Duan
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Darlynn M. Rojo-Wissar
- The Initiative on Stress, Trauma, and Resilience (STAR), Department of Psychiatry and Human Behavior, Center for Behavioral and Preventive Medicine, The Miriam Hospital, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Calliope Holingue
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Center for Autism and Related Disorders, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Adam P. Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Johns Hopkins Center on Aging and Health, Baltimore, MD, United States
| |
Collapse
|
11
|
Ghosh-Swaby OR, Reichelt AC, Sheppard PAS, Davies J, Bussey TJ, Saksida LM. Metabolic hormones mediate cognition. Front Neuroendocrinol 2022; 66:101009. [PMID: 35679900 DOI: 10.1016/j.yfrne.2022.101009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/18/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
Abstract
Recent biochemical and behavioural evidence indicates that metabolic hormones not only regulate energy intake and nutrient content, but also modulate plasticity and cognition in the central nervous system. Disruptions in metabolic hormone signalling may provide a link between metabolic syndromes like obesity and diabetes, and cognitive impairment. For example, altered metabolic homeostasis in obesity is a strong determinant of the severity of age-related cognitive decline and neurodegenerative disease. Here we review the evidence that eating behaviours and metabolic hormones-particularly ghrelin, leptin, and insulin-are key players in the delicate regulation of neural plasticity and cognition. Caloric restriction and antidiabetic therapies, both of which affect metabolic hormone levels can restore metabolic homeostasis and enhance cognitive function. Thus, metabolic hormone pathways provide a promising target for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Olivia R Ghosh-Swaby
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada
| | - Amy C Reichelt
- Faculty of Health and Medical Sciences, Adelaide Medical School, Adelaide, Australia
| | - Paul A S Sheppard
- Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Jeffrey Davies
- Swansea University Medical School, Swansea University, Swansea, UK
| | - Timothy J Bussey
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Lisa M Saksida
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada.
| |
Collapse
|
12
|
Tomm RJ, Seib DR, Kachkovski GV, Schweitzer HR, Tobiansky DJ, Floresco SB, Soma KK. Androgen synthesis inhibition increases behavioural flexibility and mPFC tyrosine hydroxylase in gonadectomized male rats. J Neuroendocrinol 2022; 34:e13128. [PMID: 35583989 DOI: 10.1111/jne.13128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 02/15/2022] [Accepted: 03/11/2022] [Indexed: 11/26/2022]
Abstract
Behavioural flexibility is essential to adapt to a changing environment and depends on the medial prefrontal cortex (mPFC). Testosterone administration decreases behavioural flexibility. It is well known that testosterone is produced in the gonads, but testosterone is also produced in the brain, including the mPFC and other nodes of the mesocorticolimbic system. It is unclear how testosterone produced in the brain versus the gonads influences behavioural flexibility. Here, in adult male rats, we assessed the effects of the androgen synthesis inhibitor abiraterone acetate (ABI) and long-term gonadectomy (GDX) on behavioural flexibility in two paradigms. In Experiment 1, ABI but not GDX reduced the number of errors to criterion and perseverative errors in a strategy set-shifting task. In Experiment 2, with a separate cohort of rats, ABI but not GDX reduced perseverative errors in a reversal learning task. In Experiment 1, we also examined tyrosine hydroxylase immunoreactivity (TH-ir), and ABI but not GDX increased TH-ir in the mPFC. Our findings suggest that neurally-produced androgens modulate behavioural flexibility via modification of dopamine signalling in the mesocorticolimbic system. These results indicate that neurosteroids regulate executive functions and that ABI treatment for prostate cancer might affect cognition.
Collapse
Affiliation(s)
- Ryan J Tomm
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Désirée R Seib
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - George V Kachkovski
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Helen R Schweitzer
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Daniel J Tobiansky
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Kiran K Soma
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| |
Collapse
|
13
|
He Z. Selective effects of perinatal estrogen on proliferation and new neurons in hippocampus and piriform cortex of rats at weaning. Neurotoxicology 2022; 91:254-261. [PMID: 35618077 DOI: 10.1016/j.neuro.2022.05.012] [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: 11/16/2021] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND A recent report links heightened prenatal amniotic estrogen levels to an increased risk of autism spectrum disorder (ASD). In this study, we examined the developmental effects of perinatal estrogen treatment on stem cell activity in weaned rats. METHODS Sprague-Dawley rats received ethinyl estradiol (EE2, 10µg/kg/day) or vehicle orally from gestational day 6 until parturition. Offspring were then treated with the same daily dose from postnatal days (PNDs) 1-21. The effects of perinatal estrogen treatment on stem cell activities in the subgranular zone (SGZ) of the hippocampus and the piriform cortex were evaluated in male and female rat pups. RESULTS EE2 treatment increased the total Ki67-immunoreactive (Ki67-ir) cell counts in the SGZ of males and females (p<0.05). However, no treatment or sex differences were detectable in the density of the doublecortin (DCX)-immunoreactive (DCX-ir) deposits in the hippocampus. In the piriform cortex, no treatment or sex differences were detected in Ki67-ir cell counts. However, the EE2 treatment significantly reduced the DCX-ir cell count in male, but not female rats (male EE2 group=292±22/mm2, male vehicle group=402±19/mm2, female EE2 group=342±15/mm2, female vehicle group=331±9/mm2). CONCLUSIONS Perinatal estrogen treatment increased hippocampal Ki67-ir cell counts in both sexes and selectively reduced DCX-ir cell counts in the piriform cortex of males. These data suggest that exposure to abnormally high levels of estrogens early in life may have an impact on neural cell development. Alterations in development so early in life may have long-term cognitive impact.
Collapse
Affiliation(s)
- Z He
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR 72079 USA.
| |
Collapse
|
14
|
Effects of circulating estradiol on physiological, behavioural, and subjective correlates of anxiety: A double-blind, randomized, placebo-controlled trial. Psychoneuroendocrinology 2022; 138:105682. [PMID: 35123210 DOI: 10.1016/j.psyneuen.2022.105682] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 12/17/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022]
Abstract
Anxiety-related behaviours as well as the prevalence of anxiety disorders show a large sex difference in humans. Clinical studies in humans as well as behavioural studies in rodents suggest that estradiol may have anxiolytic properties. In line with this, anxiety symptoms fluctuate with estradiol levels along the menstrual cycle. However, the influence of estradiol on subjective, behavioural, as well as physiological correlates of anxiety has never been systematically addressed in humans. We ran a double-blind, randomized, placebo-controlled study (N = 126) to investigate the effects of estradiol on anxiety in men and women. In healthy volunteers, circulating estradiol levels were elevated through estradiol administration over two consecutive days to simulate the rise in estradiol levels around ovulation. Subjective, behavioral, as well as, physiological correlates of anxiety were assessed using a virtual reality elevated plus-maze (EPM). Estradiol treatment reduced the physiological stress response with blunted heart rate response and lower cortisol levels compared to placebo treatment in both sexes. In contrast, respiration frequency was only reduced in women after estradiol treatment. Behavioural measures of anxiety as well as subjective anxiety on the EPM were not affected by estradiol treatment. In general, women showed more avoidance and less approach behavior and reported higher subjective anxiety levels on the EPM than men. These results highlight the limited anxiolytic properties of circulating levels of estradiol in humans, which influence physiological markers of anxiety but not approach and avoidance behaviour or subjective anxiety levels.
Collapse
|
15
|
Grigoryan GA. Ovariectomy as a Model of Anxiety-Depressive Disorders. NEUROCHEM J+ 2022. [DOI: 10.1134/s1819712422010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Rabin RA, Mackey S, Parvaz MA, Cousijn J, Li C, Pearlson G, Schmaal L, Sinha R, Stein E, Veltman D, Thompson PM, Conrod P, Garavan H, Alia‐Klein N, Goldstein RZ. Common and gender-specific associations with cocaine use on gray matter volume: Data from the ENIGMA addiction working group. Hum Brain Mapp 2022; 43:543-554. [PMID: 32857473 PMCID: PMC8675419 DOI: 10.1002/hbm.25141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/26/2020] [Accepted: 07/10/2020] [Indexed: 11/30/2022] Open
Abstract
Gray matter volume (GMV) in frontal cortical and limbic regions is susceptible to cocaine-associated reductions in cocaine-dependent individuals (CD) and is negatively associated with duration of cocaine use. Gender differences in CD individuals have been reported clinically and in the context of neural responses to cue-induced craving and stress reactivity. The variability of GMV in select brain areas between men and women (e.g., limbic regions) underscores the importance of exploring interaction effects between gender and cocaine dependence on brain structure. Therefore, voxel-based morphometry data derived from the ENIGMA Addiction Consortium were used to investigate potential gender differences in GMV in CD individuals compared to matched controls (CTL). T1-weighted MRI scans and clinical data were pooled from seven sites yielding 420 gender- and age-matched participants: CD men (CDM, n = 140); CD women (CDW, n = 70); control men (CTLM, n = 140); and control women (CTLW, n = 70). Differences in GMV were assessed using a 2 × 2 ANCOVA, and voxelwise whole-brain linear regressions were conducted to explore relationships between GMV and duration of cocaine use. All analyses were corrected for age, total intracranial volume, and site. Diagnostic differences were predominantly found in frontal regions (CD < CTL). Interestingly, gender × diagnosis interactions in the left anterior insula and left lingual gyrus were also documented, driven by differences in women (CDW < CTLW). Further, lower right hippocampal GMV was associated with greater cocaine duration in CDM. Given the importance of the anterior insula to interoception and the hippocampus to learning contextual associations, results may point to gender-specific mechanisms in cocaine addiction.
Collapse
Affiliation(s)
- Rachel A. Rabin
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Scott Mackey
- Departments of Psychiatry and PsychologyUniversity of VermontBurlingtonVermontUSA
| | - Muhammad A. Parvaz
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Janna Cousijn
- Department of PsychologyUniversity of AmsterdamAmsterdamThe Netherlands
| | - Chiang‐shan Li
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Godfrey Pearlson
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Lianne Schmaal
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Australia and Centre for Youth Mental HealthThe University of MelbourneMelbourneAustralia
| | - Rajita Sinha
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Elliot Stein
- Intramural Research Program—Neuroimaging Research BranchNational Institute on Drug AbuseBaltimoreMarylandUSA
| | - Dick Veltman
- Department of PsychiatryVU University Medical CenterAmsterdamThe Netherlands
| | - Paul M. Thompson
- Imaging Genetics Center, Department of Neurology Keck School of MedicineUniversity of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Patricia Conrod
- Department of PsychiatryUniversité de Montreal, CHU Ste Justine HospitalMontrealQuebecCanada
| | - Hugh Garavan
- Departments of Psychiatry and PsychologyUniversity of VermontBurlingtonVermontUSA
| | - Nelly Alia‐Klein
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Rita Z. Goldstein
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
17
|
Noorjahan N, Cattini PA. Neurogenesis in the Maternal Rodent Brain: Impacts of Gestation-Related Hormonal Regulation, Stress, and Obesity. Neuroendocrinology 2022; 112:702-722. [PMID: 34510034 DOI: 10.1159/000519415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 09/02/2021] [Indexed: 11/19/2022]
Abstract
In order to maintain maternal behavior, it is important that the maternal rodent brain promotes neurogenesis. Maternal neurogenesis is altered by the dynamic shifts in reproductive hormone levels during pregnancy. Thus, lifestyle events such as gestational stress and obesity that can affect hormone production will affect neuroendocrine control of maternal neurogenesis. However, there is a lack of information about the regulation of maternal neurogenesis by placental hormones, which are key components of the reproductive hormonal profile during pregnancy. There is also little known about how maternal neurogenesis can be affected by health concerns such as gestational stress and obesity, and its relationship to peripartum mental health disorders. This review summarizes the changing levels of neurogenesis in mice and rats during gestation and postpartum as well as regulation of neurogenesis by pregnancy-related hormones. The influence of neurogenesis on maternal behavior is also discussed while bringing attention to the effect of health-related concerns during gestation, such as stress and obesity on neuroendocrine control of maternal neurogenesis. In doing so, this review identifies the gaps in the literature and specifically emphasizes the importance of further research on maternal brain physiology to address them.
Collapse
Affiliation(s)
- Noshin Noorjahan
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter A Cattini
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
18
|
Kuwahara N, Nicholson K, Isaacs L, MacLusky NJ. Androgen Effects on Neural Plasticity. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:216-230. [PMID: 35024693 PMCID: PMC8744448 DOI: 10.1089/andro.2021.0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Androgens are synthesized in the brain, gonads, and adrenal glands, in both sexes, exerting physiologically important effects on the structure and function of the central nervous system. These effects may contribute to the incidence and progression of neurological disorders such as autism spectrum disorder, schizophrenia, and Alzheimer's disease, which occur at different rates in males and females. This review briefly summarizes the current state of knowledge with respect to the neuroplastic effects of androgens, with particular emphasis on the hippocampus, which has been the focus of much of the research in this field.
Collapse
Affiliation(s)
- Nariko Kuwahara
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kate Nicholson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Lauren Isaacs
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Neil J. MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
19
|
Hodges TE, Puri TA, Blankers SA, Qiu W, Galea LAM. Steroid hormones and hippocampal neurogenesis in the adult mammalian brain. VITAMINS AND HORMONES 2021; 118:129-170. [PMID: 35180925 DOI: 10.1016/bs.vh.2021.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hippocampal neurogenesis persists across the lifespan in many species, including rodents and humans, and is associated with cognitive performance and the pathogenesis of neurodegenerative disease and psychiatric disorders. Neurogenesis is modulated by steroid hormones that change across development and differ between the sexes in rodents and humans. Here, we discuss the effects of stress and glucocorticoid exposure from gestation to adulthood as well as the effects of androgens and estrogens in adulthood on neurogenesis in the hippocampus. Throughout the review we highlight sex differences in the effects of steroid hormones on neurogenesis and how they may relate to hippocampal function and disease. These data highlight the importance of examining age and sex when evaluating the effects of steroid hormones on hippocampal neurogenesis.
Collapse
Affiliation(s)
- Travis E Hodges
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
20
|
Blankers SA, Galea LA. Androgens and Adult Neurogenesis in the Hippocampus. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:203-215. [PMID: 35024692 PMCID: PMC8744005 DOI: 10.1089/andro.2021.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 11/12/2022]
Abstract
Adult neurogenesis in the hippocampus is modulated by steroid hormones, including androgens, in male rodents. In this review, we summarize research showing that chronic exposure to androgens, such as testosterone and dihydrotestosterone, enhances the survival of new neurons in the dentate gyrus of male, but not female, rodents, via the androgen receptor. However, the neurogenesis promoting the effect of androgens in the dentate gyrus may be limited to younger adulthood as it is not evident in middle-aged male rodents. Although direct exposure to androgens in adult or middle age does not significantly influence neurogenesis in female rodents, the aromatase inhibitor letrozole enhances neurogenesis in the hippocampus of middle-aged female mice. Unlike other androgens, androgenic anabolic steroids reduce neurogenesis in the hippocampus of male rodents. Collectively, the research indicates that the ability of androgens to enhance hippocampal neurogenesis in adult rodents is dependent on dose, androgen type, sex, duration, and age. We discuss these findings and how androgens may be influencing neuroprotection, via neurogenesis in the hippocampus, in the context of health and disease.
Collapse
Affiliation(s)
- Samantha A. Blankers
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
| | - Liisa A.M. Galea
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
- Department of Psychology, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
21
|
Nawarawong NN, Thompson KR, Guerin SP, Anasooya Shaji C, Peng H, Nixon K. Reactive, Adult Neurogenesis From Increased Neural Progenitor Cell Proliferation Following Alcohol Dependence in Female Rats. Front Neurosci 2021; 15:689601. [PMID: 34594180 PMCID: PMC8477003 DOI: 10.3389/fnins.2021.689601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/16/2021] [Indexed: 11/25/2022] Open
Abstract
Hippocampal neurodegeneration is a consequence of excessive alcohol drinking in alcohol use disorders (AUDs), however, recent studies suggest that females may be more susceptible to alcohol-induced brain damage. Adult hippocampal neurogenesis is now well accepted to contribute to hippocampal integrity and is known to be affected by alcohol in humans as well as in animal models of AUDs. In male rats, a reactive increase in adult hippocampal neurogenesis has been observed during abstinence from alcohol dependence, a phenomenon that may underlie recovery of hippocampal structure and function. It is unknown whether reactive neurogenesis occurs in females. Therefore, adult female rats were exposed to a 4-day binge model of alcohol dependence followed by 7 or 14 days of abstinence. Immunohistochemistry (IHC) was used to assess neural progenitor cell (NPC) proliferation (BrdU and Ki67), the percentage of increased NPC activation (Sox2+/Ki67+), the number of immature neurons (NeuroD1), and ectopic dentate gyrus granule cells (Prox1). On day seven of abstinence, ethanol-treated females showed a significant increase in BrdU+ and Ki67+ cells in the subgranular zone of the dentate gyrus (SGZ), as well as greater activation of NPCs (Sox2+/Ki67+) into active cycling. At day 14 of abstinence, there was a significant increase in the number of immature neurons (NeuroD1+) though no evidence of ectopic neurogenesis according to either NeuroD1 or Prox1 immunoreactivity. Altogether, these data suggest that alcohol dependence produces similar reactive increases in NPC proliferation and adult neurogenesis. Thus, reactive, adult neurogenesis may be a means of recovery for the hippocampus after alcohol dependence in females.
Collapse
Affiliation(s)
- Natalie N Nawarawong
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - K Ryan Thompson
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Steven P Guerin
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | | | - Hui Peng
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Kimberly Nixon
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
22
|
Wan L, Huang RJ, Luo ZH, Gong JE, Pan A, Manavis J, Yan XX, Xiao B. Reproduction-Associated Hormones and Adult Hippocampal Neurogenesis. Neural Plast 2021; 2021:3651735. [PMID: 34539776 PMCID: PMC8448607 DOI: 10.1155/2021/3651735] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/17/2021] [Indexed: 11/18/2022] Open
Abstract
The levels of reproduction-associated hormones in females, such as estrogen, progesterone, prolactin, and oxytocin, change dramatically during pregnancy and postpartum. Reproduction-associated hormones can affect adult hippocampal neurogenesis (AHN), thereby regulating mothers' behavior after delivery. In this review, we first briefly introduce the overall functional significance of AHN and the methods commonly used to explore this front. Then, we attempt to reconcile the changes of reproduction-associated hormones during pregnancy. We further update the findings on how reproduction-related hormones influence adult hippocampal neurogenesis. This review is aimed at emphasizing a potential role of AHN in reproduction-related brain plasticity and its neurobiological relevance to motherhood behavior.
Collapse
Affiliation(s)
- Lily Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Rou-Jie Huang
- Medical Doctor Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhao-Hui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jiao-e Gong
- Department of Neurology, Hunan Children's Hospital, Changsha 410007, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, Hunan 410013, China
| | - Jim Manavis
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia 5000
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, Hunan 410013, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
23
|
Akhter F, Persaud A, Zaokari Y, Zhao Z, Zhu D. Vascular Dementia and Underlying Sex Differences. Front Aging Neurosci 2021; 13:720715. [PMID: 34566624 PMCID: PMC8457333 DOI: 10.3389/fnagi.2021.720715] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/13/2021] [Indexed: 11/29/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia after Alzheimer's disease (AD); where Alzheimer's accounts for 60-70% of cases of dementia and VaD accounts for 20% of all dementia cases. VaD is defined as a reduced or lack of blood flow to the brain that causes dementia. VaD is also known occasionally as vascular contributions to cognitive impairment and dementia (VCID) or multi-infarct dementia (MID). VCID is the condition arising from stroke and other vascular brain injuries that cause significant changes to memory, thinking, and behavior, and VaD is the most severe stage while MID is produced by the synergistic effects caused by multiple mini strokes in the brain irrespective of specific location or volume. There are also subtle differences in the presentation of VaD in males and females, but they are often overlooked. Since 1672 when the first case of VaD was reported until now, sex and gender differences have had little to no research done when it comes to the umbrella term of dementia in general. This review summarizes the fundamentals of VaD followed by a focus on the differences between sex and gender when an individual is diagnosed. In addition, we provide critical evidence concerning sex and gender differences with a few of the main risk factors of VaD including pre-existing health conditions and family history, gene variants, aging, hormone fluctuations, and environmental risk factors. Additionally, the pharmaceutical treatments and possible mitigation of risk factors is explored.
Collapse
Affiliation(s)
- Firoz Akhter
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Alicia Persaud
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Younis Zaokari
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Zhen Zhao
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
- Neuroscience Graduate Program, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
24
|
Kawatake-Kuno A, Murai T, Uchida S. The Molecular Basis of Depression: Implications of Sex-Related Differences in Epigenetic Regulation. Front Mol Neurosci 2021; 14:708004. [PMID: 34276306 PMCID: PMC8282210 DOI: 10.3389/fnmol.2021.708004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/14/2021] [Indexed: 12/22/2022] Open
Abstract
Major depressive disorder (MDD) is a leading cause of disability worldwide. Although the etiology and pathophysiology of MDD remain poorly understood, aberrant neuroplasticity mediated by the epigenetic dysregulation of gene expression within the brain, which may occur due to genetic and environmental factors, may increase the risk of this disorder. Evidence has also been reported for sex-related differences in the pathophysiology of MDD, with female patients showing a greater severity of symptoms, higher degree of functional impairment, and more atypical depressive symptoms. Males and females also differ in their responsiveness to antidepressants. These clinical findings suggest that sex-dependent molecular and neural mechanisms may underlie the development of depression and the actions of antidepressant medications. This review discusses recent advances regarding the role of epigenetics in stress and depression. The first section presents a brief introduction of the basic mechanisms of epigenetic regulation, including histone modifications, DNA methylation, and non-coding RNAs. The second section reviews their contributions to neural plasticity, the risk of depression, and resilience against depression, with a particular focus on epigenetic modulators that have causal relationships with stress and depression in both clinical and animal studies. The third section highlights studies exploring sex-dependent epigenetic alterations associated with susceptibility to stress and depression. Finally, we discuss future directions to understand the etiology and pathophysiology of MDD, which would contribute to optimized and personalized therapy.
Collapse
Affiliation(s)
- Ayako Kawatake-Kuno
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshiya Murai
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shusaku Uchida
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
25
|
Li Q, Lou J, Yang T, Wei Z, Li S, Zhang F. Ischemic Preconditioning Induces Oligodendrogenesis in Mouse Brain: Effects of Nrf2 Deficiency. Cell Mol Neurobiol 2021; 42:1859-1873. [PMID: 33666795 DOI: 10.1007/s10571-021-01068-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/23/2021] [Indexed: 10/22/2022]
Abstract
Ischemic preconditioning (IPC) is an approach of protection against cerebral ischemia by inducing endogenous cytoprotective machinery. However, few studies in neurogenesis and oligodendrogenesis after IPC have been reported, especially the latter. The purpose of this study is to test our hypothesis that IPC may also induce cell proliferation and oligodendrogenesis in the subventricular zone and striatum, as well as to investigate the effect of nuclear factor erythroid 2-related factor 2 (Nrf2) on oligodendrogenesis. IPC was induced in mice by 12-min ischemia through the occlusion of the middle cerebral artery. Newly generated cells were labeled with 5-bromo-2'-deoxyuridine. Our findings demonstrated that IPC stimulated the proliferation of neural stem cells in the subventricular zone, promoted the generation of oligodendrocyte precursor cells in the striatum and corpus callosum/external capsule (CC/EC), and stimulated oligodendrocyte precursor cells differentiation into oligodendrocytes in the striatum and the CC/EC. Furthermore, we describe a crucial role for Nrf2 in IPC-induced oligodendrogenesis in the subventricular zone, striatum, and CC/EC and show for the first time that Nrf2 promoted the migration and differentiation of oligodendrocyte precursor cells into oligodendrocytes in the striatum and CC/EC. Our data imply that IPC stimulates the oligodendrogenesis in the brain and that Nrf2 signaling may contribute to the oligodendrogenesis.
Collapse
Affiliation(s)
- Qianqian Li
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jiyu Lou
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tuo Yang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zhishuo Wei
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Senmiao Li
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Feng Zhang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
26
|
Trova S, Bovetti S, Pellegrino G, Bonzano S, Giacobini P, Peretto P. HPG-Dependent Peri-Pubertal Regulation of Adult Neurogenesis in Mice. Front Neuroanat 2020; 14:584493. [PMID: 33328903 PMCID: PMC7732626 DOI: 10.3389/fnana.2020.584493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/29/2020] [Indexed: 11/13/2022] Open
Abstract
Adult neurogenesis, a striking form of neural plasticity, is involved in the modulation of social stimuli driving reproduction. Previous studies on adult neurogenesis have shown that this process is significantly modulated around puberty in female mice. Puberty is a critical developmental period triggered by increased secretion of the gonadotropin releasing hormone (GnRH), which controls the activity of the hypothalamic-pituitary-gonadal axis (HPG). Secretion of HPG-axis factors at puberty participates to the refinement of neural circuits that govern reproduction. Here, by exploiting a transgenic GnRH deficient mouse model, that progressively loses GnRH expression during postnatal development (GnRH::Cre;Dicer loxP/loxP mice), we found that a postnatally-acquired dysfunction in the GnRH system affects adult neurogenesis selectively in the subventricular-zone neurogenic niche in a sexually dimorphic way. Moreover, by examining adult females ovariectomized before the onset of puberty, we provide important evidence that, among the HPG-axis secreting factors, the circulating levels of gonadal hormones during pre-/peri-pubertal life contribute to set-up the proper adult subventricular zone-olfactory bulb neurogenic system.
Collapse
Affiliation(s)
- Sara Trova
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy.,Univ.Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, Laboratory of the Development and Plasticity of Neuroendocrine Brain, Lille, France
| | - Serena Bovetti
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| | - Giuliana Pellegrino
- Univ.Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, Laboratory of the Development and Plasticity of Neuroendocrine Brain, Lille, France
| | - Sara Bonzano
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| | - Paolo Giacobini
- Univ.Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, Laboratory of the Development and Plasticity of Neuroendocrine Brain, Lille, France
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| |
Collapse
|
27
|
Premachandran H, Zhao M, Arruda-Carvalho M. Sex Differences in the Development of the Rodent Corticolimbic System. Front Neurosci 2020; 14:583477. [PMID: 33100964 PMCID: PMC7554619 DOI: 10.3389/fnins.2020.583477] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
In recent years, a growing body of research has shown sex differences in the prevalence and symptomatology of psychopathologies, such as depression, anxiety, and fear-related disorders, all of which show high incidence rates in early life. This has highlighted the importance of including female subjects in animal studies, as well as delineating sex differences in neural processing across development. Of particular interest is the corticolimbic system, comprising the hippocampus, amygdala, and medial prefrontal cortex. In rodents, these corticolimbic regions undergo dynamic changes in early life, and disruption to their normative development is believed to underlie the age and sex-dependent effects of stress on affective processing. In this review, we consolidate research on sex differences in the hippocampus, amygdala, and medial prefrontal cortex across early development. First, we briefly introduce current principles on sexual differentiation of the rodent brain. We then showcase corticolimbic regional sex differences in volume, morphology, synaptic organization, cell proliferation, microglia, and GABAergic signaling, and explain how these differences are influenced by perinatal and pubertal gonadal hormones. In compiling this research, we outline evidence of what and when sex differences emerge in the developing corticolimbic system, and illustrate how temporal dynamics of its maturational trajectory may differ in male and female rodents. This will help provide insight into potential neural mechanisms underlying sex-specific critical windows for stress susceptibility and behavioral emergence.
Collapse
Affiliation(s)
| | - Mudi Zhao
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada.,Department of Cell and Systems Biology, University of Toronto Scarborough, Toronto, ON, Canada
| |
Collapse
|
28
|
Jorgensen C, Wang Z. Hormonal Regulation of Mammalian Adult Neurogenesis: A Multifaceted Mechanism. Biomolecules 2020; 10:biom10081151. [PMID: 32781670 PMCID: PMC7465680 DOI: 10.3390/biom10081151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023] Open
Abstract
Adult neurogenesis—resulting in adult-generated functioning, integrated neurons—is still one of the most captivating research areas of neuroplasticity. The addition of new neurons in adulthood follows a seemingly consistent multi-step process. These neurogenic stages include proliferation, differentiation, migration, maturation/survival, and integration of new neurons into the existing neuronal network. Most studies assessing the impact of exogenous (e.g., restraint stress) or endogenous (e.g., neurotrophins) factors on adult neurogenesis have focused on proliferation, survival, and neuronal differentiation. This review will discuss the multifaceted impact of hormones on these various stages of adult neurogenesis. Specifically, we will review the evidence for hormonal facilitation (via gonadal hormones), inhibition (via glucocorticoids), and neuroprotection (via recruitment of other neurochemicals such as neurotrophin and neuromodulators) on newly adult-generated neurons in the mammalian brain.
Collapse
Affiliation(s)
- Claudia Jorgensen
- Behavioral Science Department, Utah Valley University, Orem, UT 84058, USA
- Correspondence:
| | - Zuoxin Wang
- Psychology Department and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| |
Collapse
|
29
|
Bettio LEB, Thacker JS, Rodgers SP, Brocardo PS, Christie BR, Gil-Mohapel J. Interplay between hormones and exercise on hippocampal plasticity across the lifespan. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165821. [PMID: 32376385 DOI: 10.1016/j.bbadis.2020.165821] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/19/2020] [Accepted: 04/25/2020] [Indexed: 12/15/2022]
Abstract
The hippocampus is a brain structure known to play a central role in cognitive function (namely learning and memory) as well as mood regulation and affective behaviors due in part to its ability to undergo structural and functional changes in response to intrinsic and extrinsic stimuli. While structural changes are achieved through modulation of hippocampal neurogenesis as well as alterations in dendritic morphology and spine remodeling, functional (i.e., synaptic) changes can be noted through the strengthening (i.e., long-term potentiation) or weakening (i.e., long-term depression) of the synapses. While age, hormone homeostasis, and levels of physical activity are some of the factors known to module these forms of hippocampal plasticity, the exact mechanisms through which these factors interact with each other at a given moment in time are not completely understood. It is well known that hormonal levels vary throughout the lifespan of an individual and it is also known that physical exercise can impact hormonal homeostasis. Thus, it is reasonable to speculate that hormone modulation might be one of the various mechanisms through which physical exercise differently impacts hippocampal plasticity throughout distinct periods of an individual's life. The present review summarizes the potential relationship between physical exercise and different types of hormones (namely sex, metabolic, and stress hormones) and how this relationship may mediate the effects of physical activity during three distinct life periods, adolescence, adulthood, and senescence. Overall, the vast majority of studies support a beneficial role of exercise in maintaining hippocampal hormonal levels and consequently, hippocampal plasticity, cognition, and mood regulation.
Collapse
Affiliation(s)
- Luis E B Bettio
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Jonathan S Thacker
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Shaefali P Rodgers
- Developmental, Cognitive & Behavioral Neuroscience Program, Department of Psychology, Texas Institute for Measurement, Evaluation, and Statistics, University of Houston, TX, USA
| | - Patricia S Brocardo
- Department of Morphological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Brian R Christie
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada; Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC, Canada
| | - Joana Gil-Mohapel
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada; Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC, Canada.
| |
Collapse
|
30
|
Sex Differences in Maturation and Attrition of Adult Neurogenesis in the Hippocampus. eNeuro 2020; 7:ENEURO.0468-19.2020. [PMID: 32586842 PMCID: PMC7369314 DOI: 10.1523/eneuro.0468-19.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Sex differences exist in the regulation of adult neurogenesis in the hippocampus in response to hormones and cognitive training. Here, we investigated the trajectory and maturation rate of adult-born neurons in the dentate gyrus (DG) of male and female rats. Sprague Dawley rats were perfused 2 h, 24 h, one week (1w), 2w, or 3w after bromodeoxyuridine (BrdU) injection, a DNA synthesis marker that labels dividing progenitor cells and their progeny. Adult-born neurons (BrdU/NeuN-ir) matured faster in males compared with females. Males had a greater density of neural stem cells (Sox2-ir) in the dorsal, but not in the ventral, DG and had higher levels of cell proliferation (Ki67-ir) than non-proestrous females. However, males showed a greater reduction in neurogenesis between 1week and 2weeks after mitosis, whereas females showed similar levels of neurogenesis throughout the weeks. The faster maturation and greater attrition of new neurons in males compared with females suggests greater potential for neurogenesis to respond to external stimuli in males and emphasizes the importance of studying sex on adult hippocampal neurogenesis.
Collapse
|
31
|
Duarte-Guterman P, Lieblich SE, Qiu W, Splinter JEJ, Go KA, Casanueva-Reimon L, Galea LAM. Oxytocin has sex-specific effects on social behaviour and hypothalamic oxytocin immunoreactive cells but not hippocampal neurogenesis in adult rats. Horm Behav 2020; 122:104734. [PMID: 32169583 DOI: 10.1016/j.yhbeh.2020.104734] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/06/2020] [Accepted: 02/28/2020] [Indexed: 10/24/2022]
Abstract
Oxytocin regulates social behaviours, pair bonding and hippocampal neurogenesis but most studies have used adult males. Our study investigated the effects of oxytocin on social investigation and adult hippocampal neurogenesis in male and female rats. Oxytocin has poor penetration of the blood-brain barrier, therefore we tested a nanoparticle drug, TRIOZAN™ (Ovensa Inc.), which permits greater blood-brain-barrier penetration. Adult male and female rats were injected daily (i.p.) for 10 days with either: oxytocin in PBS (0.5 or 1.0 mg/kg), oxytocin in TRIOZAN™ (0.5 or 1.0 mg/kg), or vehicle (PBS) and tested for social investigation. Oxytocin decreased body mass and increased social investigation and number of oxytocin-immunoreactive cells in the supraoptic nucleus (SON) of the hypothalamus in male rats only. In both sexes, oxytocin decreased the number of immature neurons (doublecortin+ cells) in the ventral hippocampus and reduced plasma 17β-estradiol levels in a dose- and delivery-dependent way. Oxytocin in TRIOZAN™ reduced "sedation" observed post-injection and increased certain central effects (oxytocin levels in the hypothalamus and neurogenesis in the ventral hippocampus) relative to oxytocin in PBS, indicating that the nanoparticle may be used as an alternative brain delivery system. We showed that oxytocin has sex-specific effects on social investigation, body mass, "sedation", and the oxytocin system. In contrast, similar effects were observed in both sexes in neurogenesis and plasma 17β-estradiol. Our work suggests that sex differences in oxytocin regulation of brain endpoints is region-specific (hypothalamus versus hippocampus) and that oxytocin does not promote social investigation in females.
Collapse
Affiliation(s)
- Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Jared E J Splinter
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Kimberly A Go
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Laura Casanueva-Reimon
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
32
|
Christensen A, Liu J, Pike CJ. Aging Reduces Estradiol Protection Against Neural but Not Metabolic Effects of Obesity in Female 3xTg-AD Mice. Front Aging Neurosci 2020; 12:113. [PMID: 32431604 PMCID: PMC7214793 DOI: 10.3389/fnagi.2020.00113] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/03/2020] [Indexed: 12/29/2022] Open
Abstract
Vulnerability to Alzheimer's disease (AD) is increased by several risk factors, including midlife obesity, female sex, and the depletion of estrogens in women as a consequence of menopause. Conversely, estrogen-based hormone therapies have been linked with protection from age-related increases in adiposity and dementia risk, although treatment efficacy appears to be affected by the age of initiation. Potential interactions between obesity, AD, aging, and estrogen treatment are likely to have significant impact on optimizing the use of hormone therapies in postmenopausal women. In the current study, we compared how treatment with the primary estrogen, 17β-estradiol (E2), affects levels of AD-like neuropathology, behavioral impairment, and other neural and systemic effects of preexisting diet-induced obesity in female 3xTg-AD mice. Importantly, experiments were conducted at chronological ages associated with both the early and late stages of reproductive senescence. We observed that E2 treatment was generally associated with significantly improved metabolic outcomes, including reductions in body weight, adiposity, and leptin, across both age groups. Conversely, neural benefits of E2 in obese mice, including decreased β-amyloid burden, improved behavioral performance, and reduced microglial activation, were observed only in the early aging group. These results are consistent with the perspective that neural benefits of estrogen-based therapies require initiation of treatment during early rather than later phases of reproductive aging. Further, the discordance between E2 protection against systemic versus neural effects of obesity across age groups suggests that pathways other than general metabolic function, perhaps including reduced microglial activation, contribute to the mechanism(s) of the observed E2 actions. These findings reinforce the potential systemic and neural benefits of estrogen therapies against obesity, while also highlighting the critical role of aging as a mediator of estrogens' protective actions.
Collapse
Affiliation(s)
| | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
33
|
Eid RS, Lieblich SE, Duarte-Guterman P, Chaiton JA, Mah AG, Wong SJ, Wen Y, Galea LAM. Selective activation of estrogen receptors α and β: Implications for depressive-like phenotypes in female mice exposed to chronic unpredictable stress. Horm Behav 2020; 119:104651. [PMID: 31790664 DOI: 10.1016/j.yhbeh.2019.104651] [Citation(s) in RCA: 10] [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: 09/07/2019] [Revised: 11/06/2019] [Accepted: 11/26/2019] [Indexed: 01/19/2023]
Abstract
The estrogen receptor (ER) mechanisms by which 17β-estradiol influences depressive-like behaviour have primarily been investigated acutely and not within an animal model of depression. Therefore, the current study aimed to dissect the contribution of ERα and ERβ to the effects of 17β-estradiol under non-stress and chronic stress conditions. Ovariectomized (OVX) or sham-operated mice were treated chronically (47 days) with 17β-estradiol (E2), the ERβ agonist diarylpropionitrile (DPN), the ERα agonist propylpyrazole-triol (PPT), or vehicle. On day 15 of treatment, mice from each group were assigned to chronic unpredictable stress (CUS; 28 days) or non-CUS conditions. Mice were assessed for anxiety- and depressive-like behaviour and hypothalamic-pituitary-adrenal (HPA) axis function. Cytokine and chemokine levels, and postsynaptic density protein 95 were measured in the hippocampus and frontal cortex, and adult hippocampal neurogenesis was assessed. Overall, the effects of CUS were more robust that those of estrogenic treatments, as seen by increased immobility in the tail suspension test (TST), reduced PSD-95 expression, reduced neurogenesis in the ventral hippocampus, and HPA axis negative feedback dysregulation. However, we also observe CUS-dependent and -independent effects of ovarian status and estrogenic treatments. The effects of CUS on PSD-95 expression, the cytokine milieu, and in TST were largely driven by PPT and DPN, indicating that these treatments were not protective. Independent of CUS, estradiol increased neurogenesis in the dorsal hippocampus, blunted the corticosterone response to an acute stressor, and increased anxiety-like behaviour. These findings provide insights into the complexities of estrogen signaling in modulating depressive-like phenotypes under non-stress and chronic stress conditions.
Collapse
Affiliation(s)
- Rand S Eid
- Graduate program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Paula Duarte-Guterman
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Jessica A Chaiton
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Amanda G Mah
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Sarah J Wong
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Yanhua Wen
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
34
|
Spritzer MD, Roy EA. Testosterone and Adult Neurogenesis. Biomolecules 2020; 10:biom10020225. [PMID: 32028656 PMCID: PMC7072323 DOI: 10.3390/biom10020225] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
It is now well established that neurogenesis occurs throughout adulthood in select brain regions, but the functional significance of adult neurogenesis remains unclear. There is considerable evidence that steroid hormones modulate various stages of adult neurogenesis, and this review provides a focused summary of the effects of testosterone on adult neurogenesis. Initial evidence came from field studies with birds and wild rodent populations. Subsequent experiments with laboratory rodents have tested the effects of testosterone and its steroid metabolites upon adult neurogenesis, as well as the functional consequences of induced changes in neurogenesis. These experiments have provided clear evidence that testosterone increases adult neurogenesis within the dentate gyrus region of the hippocampus through an androgen-dependent pathway. Most evidence indicates that androgens selectively enhance the survival of newly generated neurons, while having little effect on cell proliferation. Whether this is a result of androgens acting directly on receptors of new neurons remains unclear, and indirect routes involving brain-derived neurotrophic factor (BDNF) and glucocorticoids may be involved. In vitro experiments suggest that testosterone has broad-ranging neuroprotective effects, which will be briefly reviewed. A better understanding of the effects of testosterone upon adult neurogenesis could shed light on neurological diseases that show sex differences.
Collapse
Affiliation(s)
- Mark D. Spritzer
- Department of Biology, Middlebury College, Middlebury, VT 05753, USA
- Correspondence: ; Tel.: 802-443-5676
| | - Ethan A. Roy
- Graduate School of Education, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
35
|
Prolactin, Estradiol and Testosterone Differentially Impact Human Hippocampal Neurogenesis in an In Vitro Model. Neuroscience 2020; 454:15-39. [PMID: 31930958 PMCID: PMC7839971 DOI: 10.1016/j.neuroscience.2019.12.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 12/20/2022]
Abstract
Human hippocampal progenitor cells (HPCs) and tissue express classical sex hormone receptors. Prolactin does not impact human HPCs maintained in a proliferative state. Prolactin increases neuronal differentiation of human HPCs only in the short term. Estradiol and testosterone both increase the cell density of proliferating HPCs. Estradiol and testosterone have no observed effect on differentiating HPCs.
Previous studies have indicated that sex hormones such as prolactin, estradiol and testosterone may play a role in the modulation of adult hippocampal neurogenesis (AHN) in rodents and non-human primates, but so far there has been no investigation of their impact on human hippocampal neurogenesis. Here, we quantify the expression levels of the relevant receptors in human post-mortem hippocampal tissue and a human hippocampal progenitor cell (HPC) line. Secondly, we investigate how these hormones modulate hippocampal neurogenesis using a human in vitro cellular model. Human female HPCs were cultured with biologically relevant concentrations of either prolactin, estradiol or testosterone. Bromodeoxyuridine (BrdU) incorporation, immunocytochemistry (ICC) and high-throughput analyses were used to quantify markers determining cell fate after HPCs were either maintained in a proliferative state or allowed to differentiate in the presence of these hormones. In proliferating cells, estrogen and testosterone increased cell density but had no clear effect on markers of proliferation or cell death to account for this. In differentiating cells, a 3-day treatment of prolactin elicited a transient effect, whereby it increased the proportion of microtubule-associated protein 2 (MAP2)-positive and Doublecortin (DCX)-positive cells, but this effect was not apparent after 7-days. At this timepoint we instead observe a decrease in proliferation. Overall, our study demonstrates relatively minor, and possibly short-term effects of sex hormones on hippocampal neurogenesis in human cells. Further work will be needed to understand if our results differ to previous animal research due to species-specific differences, or whether it relates to limitations of our in vitro model.
Collapse
|
36
|
High-Fat Diet-Induced Obesity Causes Sex-Specific Deficits in Adult Hippocampal Neurogenesis in Mice. eNeuro 2020; 7:ENEURO.0391-19.2019. [PMID: 31871124 PMCID: PMC6946541 DOI: 10.1523/eneuro.0391-19.2019] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/26/2019] [Accepted: 12/01/2019] [Indexed: 12/13/2022] Open
Abstract
Adult hippocampal neurogenesis (AHN) is suppressed by high-fat (HF) diet and metabolic disease, including obesity and type 2 diabetes. Deficits in AHN may contribute to cognitive decline and increased risk of dementia and mood disorders, which have higher prevalence in women. However, sex differences in the effects of HF diet/metabolic disease on AHN have yet to be thoroughly investigated. Herein, male and female C57BL/6J mice were fed an HF or control (CON) diet from ∼2 to 6 months of age. After 3 months on the diet, mice were injected with 5-ethynyl-2′-deoxyuridine (EdU) then killed 4 weeks later. Cell proliferation, differentiation/maturation, and survival of new neurons in the dentate gyrus were assessed with immunofluorescence for EdU, Ki67, doublecortin (DCX), and NeuN. CON females had more proliferating cells (Ki67+) and neuroblasts/immature neurons (DCX+) compared with CON males; however, HF diet reduced these cells in females to the levels of males. Diet did not affect neurogenesis in males. Further, the numbers of proliferating cells and immature neurons were inversely correlated with both weight gain and glucose intolerance in females only. These effects were robust in the dorsal hippocampus, which supports cognitive processes. Assessment of microglia in the dentate gyrus using immunofluorescence for Iba1 and CD68 uncovered sex-specific effects of diet, which may contribute to observed differences in neurogenesis. These findings demonstrate sex-specific effects of HF diet/metabolic disease on AHN, and highlight the potential for targeting neurogenic deficits to treat cognitive decline and reduce the risk of dementia associated with these conditions, particularly in females.
Collapse
|
37
|
Fu Y, Zhang W, Tao B, Yang B, Yang D, Xie X, Liu P, Zhu Y, Zhou L, Chen T, Liu X, Yan Z. Gray Matter Differences Between Premature Pubertal Girls With and Without the Reactivation of the Hypothalamic-Pituitary-Gonadal Axis. Front Psychiatry 2020; 11:784. [PMID: 32848948 PMCID: PMC7432114 DOI: 10.3389/fpsyt.2020.00784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 07/22/2020] [Indexed: 11/13/2022] Open
Abstract
The onset of puberty and related hormones exerts significant effects on brain morphometric and psychosocial development. The biological mechanisms underlying how the reactivation of the hypothalamic-pituitary-gonadal (HPG) axis and puberty-related hormonal maturation sculpts human brain architecture remain elusive. To address this question, 105 premature pubertal girls (age 8-11 years) without menstruation underwent brain structural scanning on a 3T MR system, and the luteinizing hormone releasing hormone (LHRH) stimulation test was used to identify the reactivation of the HPG axis. Among the 105 girls, 63 were positive for HPG axis reactivation (HPG+), while the others showed negative (HPG-). Cortical thickness was calculated and compared between the two groups after adjusting for age. The brain regions showing inter-group differences were then extracted and correlated with the peak value of serum hormone after the LHRH stimulation test in entire sample. Compared to HPG- girls, HPG+ girls showed reduced cortical thickness mainly in the the right precuneus, right inferior temporal gyrus, and right superior frontal gyrus, while increased cortical thickness primarily in the left superior parietal lobe and right inferior parietal lobe. Linear-regression analysis revealed negative correlations between the cortical thickness of the right inferior parietal lobe with the peak value of FSH and the right precuneus with LH and E. These findings provide evidence to support the notion that the reactivation of HPG axis and changes of hormones during the early phase of hormonal maturation exert influences on the development of gray matter.
Collapse
Affiliation(s)
- Yuchuan Fu
- Department of Radiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenjing Zhang
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Bo Tao
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Beisheng Yang
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Di Yang
- Department of Radiology, Zhejiang Hospital, Hangzhou, China
| | - Xiaoling Xie
- Department of Radiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peining Liu
- Department of Child Health Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yaxin Zhu
- Department of Radiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lu Zhou
- Department of Radiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Chen
- Department of Radiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaozheng Liu
- Department of Radiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhihan Yan
- Department of Radiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
38
|
Schneider MA, Spritzer PM, Suh JS, Minuzzi L, Frey BN, Schwarz K, Costa AB, da Silva DC, Garcia CCG, Fontanari AMV, Anes M, Castan JU, Cunegatto FR, Picon FA, Luders E, Lobato MIR. The Link between Estradiol and Neuroplasticity in Transgender Women after Gender-Affirming Surgery: A Bimodal Hypothesis. Neuroendocrinology 2020; 110:489-500. [PMID: 31461715 DOI: 10.1159/000502977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 08/28/2019] [Indexed: 11/19/2022]
Abstract
For transgender individuals, gender-affirming surgery (GAS) and cross-sex hormone therapy (CSHT) are part of the gender transition process. Scientific evidence supporting the maintenance of CSHT after GAS-related gonadectomy is accumulating. However, few data are available on the impact of CSHT on the brain structure following hypogonadism. Thus, we aimed to investigate links between estradiol and brain cortical thickness (CTh) and cognition in 18 post-gonadectomy transgender women using a longitudinal design. For this purpose, the participants underwent a voluntary period of CSHT washout of at least 30 days, followed by estradiol re-institution for 60 days. High-resolution T1-weighted brain images, hormonal measures, working and verbal memory were collected at 2 time points: on the last day of the washout (t1) and on the last day of the 2-month CSHT period (t2). Between these 2 time points, CTh increased within the left precentral gyrus and right precuneus but decreased within the right lateral occipital cortex. However, these findings did not survive corrections of multiple comparisons. Nevertheless, there was a significant negative correlation between changes in estradiol levels and changes in CTh. This effect was evident in the left superior frontal gyrus, the left middle temporal gyrus, the right precuneus, the right superior temporal gyrus, and the right pars opercularis. Although there was an improvement in verbal memory following hypogonadism correction, we did not observe a significant relationship between changes in memory scores and CTh. Altogether, these findings suggest that there is a link between estradiol and CTh.
Collapse
Affiliation(s)
- Maiko A Schneider
- Gender Identity Program, Psychiatry Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil,
- Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, Ontario, Canada,
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada,
| | - Poli M Spritzer
- Gender Identity Program, Psychiatry Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Endocrinology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Jee Su Suh
- Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, Ontario, Canada
- Neuroscience Graduate Program, McMaster University, Hamilton, Ontario, Canada
| | - Luciano Minuzzi
- Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
- Neuroscience Graduate Program, McMaster University, Hamilton, Ontario, Canada
| | - Benicio N Frey
- Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
- Neuroscience Graduate Program, McMaster University, Hamilton, Ontario, Canada
| | - Karine Schwarz
- Gender Identity Program, Psychiatry Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Angelo B Costa
- Graduate Program in Psychology, Pontifícia Universidade do Rio Grande do Sul, Porto Alegre, Brazil
| | - Dhiordan C da Silva
- Gender Identity Program, Psychiatry Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Post-Graduation Program, Universidade Federal do Rio Grand do Sul, Porto Alegre, Brazil
| | - Claudia C G Garcia
- Gender Identity Program, Psychiatry Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Post-Graduation Program, Universidade Federal do Rio Grand do Sul, Porto Alegre, Brazil
| | - Anna M V Fontanari
- Gender Identity Program, Psychiatry Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Post-Graduation Program, Universidade Federal do Rio Grand do Sul, Porto Alegre, Brazil
| | - Mauricio Anes
- Medical Physics and Radiation Protection Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Juliana U Castan
- Psychology Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | - Felipe A Picon
- ADHD Outpatient Program, Adult Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Eileen Luders
- School of Psychology, University of Auckland, Auckland, New Zealand
- Laboratory of Neuro Imaging, School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Maria I R Lobato
- Gender Identity Program, Psychiatry Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, Ontario, Canada
- Psychiatry and Forensic Medical Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
39
|
Hillerer KM, Slattery DA, Pletzer B. Neurobiological mechanisms underlying sex-related differences in stress-related disorders: Effects of neuroactive steroids on the hippocampus. Front Neuroendocrinol 2019; 55:100796. [PMID: 31580837 PMCID: PMC7115954 DOI: 10.1016/j.yfrne.2019.100796] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don't model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.
Collapse
Affiliation(s)
- Katharina M Hillerer
- Department of Obstetrics and Gynaecology, Salzburger Landeskrankenhaus (SALK), Paracelsus Medical University (PMU), Clinical Research Center Salzburg (CRCS), Salzburg, Austria.
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Belinda Pletzer
- Department of Psychology, University of Salzburg, Salzburg, Austria; Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| |
Collapse
|
40
|
Ruddy RM, Adams KV, Morshead CM. Age- and sex-dependent effects of metformin on neural precursor cells and cognitive recovery in a model of neonatal stroke. SCIENCE ADVANCES 2019; 5:eaax1912. [PMID: 31535024 PMCID: PMC6739114 DOI: 10.1126/sciadv.aax1912] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/15/2019] [Indexed: 05/30/2023]
Abstract
Resident neural stem and progenitor cells, collectively termed neural precursor cells (NPCs), reside in a well-defined neurogenic niche in the subventricular zone (SVZ) and contribute to ongoing postnatal neurogenesis. It is well established that the NPC niche can alter the behavior of NPCs. NPC activation is a promising therapeutic strategy for brain repair. The drug metformin has been shown to activate neural stem cells, promote differentiation, and lead to functional motor recovery in a neonatal stroke model. We demonstrate that metformin-induced NPC expansion and functional recovery is sex hormone dependent. Metformin increases the size of the NPC pool in adult females, but not males, and promotes cognitive recovery in a model of brain injury in females, but not males. Our data demonstrate that metformin has age- and sex-dependent effects on NPCs that correlate with functional recovery, which has important implications for neural repair.
Collapse
Affiliation(s)
- Rebecca M. Ruddy
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Kelsey V. Adams
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Cindi M. Morshead
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
41
|
Duarte-Guterman P, Lieblich SE, Wainwright SR, Chow C, Chaiton JA, Watson NV, Galea LAM. Androgens Enhance Adult Hippocampal Neurogenesis in Males but Not Females in an Age-Dependent Manner. Endocrinology 2019; 160:2128-2136. [PMID: 31219567 PMCID: PMC6736050 DOI: 10.1210/en.2019-00114] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/24/2019] [Indexed: 01/27/2023]
Abstract
Androgens (testosterone and DHT) increase adult hippocampal neurogenesis by increasing survival of new neurons in male rats and mice via an androgen receptor pathway, but it is not known whether androgens regulate neurogenesis in female rats and whether the effect is age-dependent. We investigated the effects of DHT, a potent androgen, on neurogenesis in young adult and middle-aged male and female rats. Rats were gonadectomized and injected with the DNA synthesis marker bromodeoxyuridine (BrdU). The following day, rats began receiving daily injections of oil or DHT for 30 days. We evaluated cell proliferation (Ki67) and survival of new neurons (BrdU and BrdU/NeuN) in the hippocampus of male and female rats by using immunohistochemistry. As expected, DHT increased the number of BrdU+ cells in young males but surprisingly not in middle-aged males or in young and middle-aged females. In middle age, DHT increased the proportion of BrdU/NeuN cells, an effect driven by females. Androgen receptor expression also increased with aging in both female and male rats, which may contribute to a lack of DHT neurogenic effect in middle age. Our results indicate that DHT regulates adult hippocampal neurogenesis in a sex- and age-dependent manner.
Collapse
Affiliation(s)
- Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven R Wainwright
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carmen Chow
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jessica A Chaiton
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Neil V Watson
- Department of Psychology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Correspondence: Liisa A. M. Galea, PhD, Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada. E-mail:
| |
Collapse
|
42
|
Sex-specific neurogenic deficits and neurocognitive disorders in middle-aged HIV-1 Tg26 transgenic mice. Brain Behav Immun 2019; 80:488-499. [PMID: 30999016 PMCID: PMC6660421 DOI: 10.1016/j.bbi.2019.04.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 11/23/2022] Open
Abstract
Varying degrees of cognitive deficits affect over half of all HIV-1 infected patients. Because of antiretroviral treatment (ART) regimens, the HIV-1 patient population is increasing in age. Very few epidemiological studies have focused on sex-specific differences in HIV-1-associated neurocognitive disorders (HAND). The purpose of this study is to examine any possible differences between male and female mice in the progression of cognitive dementia during persistent low-level HIV-1 protein exposure, mimicking the typical clinical setting in the post-ART era. Eight to ten-month old HIV-1 Tg26(+/-) transgenic mice were utilized to assess for specific learning and memory modalities. Initial physiological screening and fear conditioning assessments revealed that Tg26 mice exhibited no significant differences in general behavioral function, contextual fear conditioning, or cued fear conditioning responses when compared to their wild-type (WT) littermates, regardless of sex. However, Barnes maze testing revealed significantly impaired short and long-term spatial memory in males, while females had impaired spatial learning abilities and short-term spatial memory. The potential cellular mechanism underlying these sex-specific neurocognitive deficits was explored with hippocampal neurogenic analysis. Compared to WT mice, both male and female Tg26(+/-) mice had fewer quiescent neural stem cells and neuroblasts in their hippocampi. Male Tg26(+/-) mice had a more robust reduction of the quiescent neural stem cell pool than female Tg26(+/-) mice. While female WT mice had a higher number of neural progenitor cells than male WT mice, only female Tg26(+/-) mice exhibited a robust reduction in the number of neural progenitor cells. Altogether, these results suggest that middle-aged male and female Tg26(+/-) mice manifest differing impairments in cognitive functioning and hippocampal neurogenesis. This study emphasizes the importance of understanding sex related differences in HAND pathology, which would aid in designing more optimized therapeutic regimens for the treatment of HAND.
Collapse
|
43
|
Sex steroid hormone modulation of neural stem cells: a critical review. Biol Sex Differ 2019; 10:28. [PMID: 31146782 PMCID: PMC6543604 DOI: 10.1186/s13293-019-0242-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/13/2019] [Indexed: 12/18/2022] Open
Abstract
While numerous in vivo experiments have sought to explore the effects of sex chromosome composition and sex steroid hormones on cellular proliferation and differentiation within the mammalian brain, far fewer studies as reviewed here, have explored these factors using a direct in vitro approach. Generally speaking, in vivo studies provide the gold standard to demonstrate applicable findings in regards to the role hormones play in development. However, in the case of neural stem cell (NSC) biology, there remain many unknown factors that likely contribute to observations made within the developed brain, specifically in regions where there are abundant sex steroid hormone receptors. For these reasons, using a NSC in vitro model may provide a more controlled and refined system to explore the direct effects of sex and hormone response, limiting the vast array of other influences on NSCs occurring during development and within adult cellular niches. These specific cellular models may have the ability to greatly improve the mechanistic understanding of changes occurring within the developing brain during the hormonal organization process, in addition to other modifications that may contribute to neuro-psychiatric sex-biased diseases.
Collapse
|
44
|
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: 19.2] [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
|
45
|
Ma L, Xu Y, Wang G, Li R. What do we know about sex differences in depression: A review of animal models and potential mechanisms. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:48-56. [PMID: 30165122 DOI: 10.1016/j.pnpbp.2018.08.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/22/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022]
Abstract
Clinical studies have shown that women are more susceptible to depression than men. Sex differences in depression have been associated with social, cultural, as well as biological factors. In spite of extensive preclinical studies in animal models for depression that have been used for understanding the mechanisms of the disease as well as for new drug development, a substantive lack of attention on sex-specific phenotypes in depression might mask the effect of sex on the outcome. In this review article, we summarize findings on the influence of sex on behavior in the most commonly used animal models for depression. We also discuss the potential underlying mechanisms of such sex-dependent variation in the phenotype, particularly in the neuroendocrine system.
Collapse
Affiliation(s)
- Lei Ma
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Yong Xu
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Gang Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China.
| | - Rena Li
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100012, China; Center for Hormone Advanced Science and Education, Roskamp Institute, Sarasota, FL 34243, USA.
| |
Collapse
|
46
|
Shakya R, Chongthammakun S. 17β-Estradiol attenuates the influence of chronic activated microglia on SH-SY5Y cell proliferation via canonical WNT signaling pathway. Neurosci Lett 2019; 692:174-180. [PMID: 30391546 DOI: 10.1016/j.neulet.2018.10.063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 10/21/2018] [Accepted: 10/31/2018] [Indexed: 01/22/2023]
Abstract
The decline in circulating estrogen following menopause or aging is likely to initiate chronic inflammatory disorders, leading to neurodegenerative disease. Though, WNT1 paracrine molecules are crucial in embryonic neuroblastoma cell proliferation, very less is known about its role in adult brain that is associated with estrogen as preventive therapeutic strategy. The present study evidenced for the first time that 17β-estradiol (E2), a potent form of estrogen, could compensate the chronic neuroinflammation-associated loss of neurons by upregulating canonical WNT signaling pathway. Lipopolysaccharide was used to induce inflammatory responses in microglial cell line. The increased secretion of IL-6 cytokine was confirmed as a marker of chronic microglial activation. LPS-conditioned microglial media significantly reduced the viable cells and proliferative markers, BrdU and CyclinD1 in SH-SY5Y. It also decreased the expression of canonical WNT signaling components; WNT1 and β-catenin, which were significantly rescued with pre- and co-treatment of 10 nM E2. Furthermore, estrogen antagonist ICI 182,780 abolished the E2-mediated recovery in WNT1 expression. Whereas, canonical WNT receptor antagonist, Dkk1 was able to inhibit E2-mediated recovery in the expression of downstream component, β-catenin. It suggests a promising role of canonical WNT signaling pathway in estrogen mediated prevention of neuronal cell loss under chronic neuroinflammatory condition.
Collapse
Affiliation(s)
- Rubina Shakya
- Anatomy and Structural Biology Graduate Program, Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Sukumal Chongthammakun
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
47
|
Yagi S, Galea LAM. Sex differences in hippocampal cognition and neurogenesis. Neuropsychopharmacology 2019; 44:200-213. [PMID: 30214058 PMCID: PMC6235970 DOI: 10.1038/s41386-018-0208-4] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022]
Abstract
Sex differences are reported in hippocampal plasticity, cognition, and in a number of disorders that target the integrity of the hippocampus. For example, meta-analyses reveal that males outperform females on hippocampus-dependent tasks in rodents and in humans, furthermore women are more likely to experience greater cognitive decline in Alzheimer's disease and depression, both diseases characterized by hippocampal dysfunction. The hippocampus is a highly plastic structure, important for processing higher order information and is sensitive to the environmental factors such as stress. The structure retains the ability to produce new neurons and this process plays an important role in pattern separation, proactive interference, and cognitive flexibility. Intriguingly, there are prominent sex differences in the level of neurogenesis and the activation of new neurons in response to hippocampus-dependent cognitive tasks in rodents. However, sex differences in spatial performance can be nuanced as animal studies have demonstrated that there are task, and strategy choice dependent sex differences in performance, as well as sex differences in the subregions of the hippocampus influenced by learning. This review discusses sex differences in pattern separation, pattern completion, spatial learning, and links between adult neurogenesis and these cognitive functions of the hippocampus. We emphasize the importance of including both sexes when studying genomic, cellular, and structural mechanisms of the hippocampal function.
Collapse
Affiliation(s)
- Shunya Yagi
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, 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
|
48
|
Abd-Rabo MM, Georgy GS, Saied NM, Hassan WA. Involvement of the serotonergic system and neuroplasticity in the antidepressant effect of curcumin in ovariectomized rats: Comparison with oestradiol and fluoxetine. Phytother Res 2018; 33:387-396. [DOI: 10.1002/ptr.6232] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Marwa M. Abd-Rabo
- Department of hormone; National Organization for Drug Control and Research; Giza Egypt
| | - Gehan S. Georgy
- Department of Pharmacology; National Organization for Drug Control and Research; Giza Egypt
| | - Nashwa M. Saied
- Department of hormone; National Organization for Drug Control and Research; Giza Egypt
| | - Wafaa A. Hassan
- Department of hormone; National Organization for Drug Control and Research; Giza Egypt
| |
Collapse
|
49
|
Abstract
Engaging in targeted exercise interventions is a promising, non-pharmacological strategy to mitigate the deleterious effects of aging and disease on brain health. However, despite its therapeutic potential, a large amount of variation exists in exercise efficacy in older adults aged 55 and older. In this review, we present the argument that biological sex may be an important moderator of the relationship between physical activity and cognition. Sex differences exist in dementia as well as in several associated risk factors, including genetics, cardiovascular factors, inflammation, hormones and social and psychological factors. Different exercise interventions, such as aerobic training and resistance training, influence cognition and brain health in older adults and these effects may be sex-dependent. The biological mechanisms underlying the beneficial effects of exercise on the brain may be different in males and females. Specifically, we examine sex differences in neuroplasticity, neurotrophic factors and physiological effects of exercise to highlight the possible mediators of sex differences in exercise efficacy on cognition. Future studies should address the potential sex difference in exercise efficacy if we are to develop effective, evidence-based exercise interventions to promote healthy brain aging for all individuals.
Collapse
Affiliation(s)
- Cindy K Barha
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada.,Department of Physical Therapy, University of British Columbia, Vancouver, Canada
| | - Teresa Liu-Ambrose
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada.,Department of Physical Therapy, University of British Columbia, Vancouver, Canada.,Centre for Hip Health and Mobility, Vancouver, Canada
| |
Collapse
|
50
|
Gemmel M, De Lacalle S, Mort SC, Hill LA, Charlier TD, Pawluski JL. Perinatal fluoxetine has enduring sexually differentiated effects on neurobehavioral outcomes related to social behaviors. Neuropharmacology 2018; 144:70-81. [PMID: 30326241 DOI: 10.1016/j.neuropharm.2018.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/02/2018] [Accepted: 10/11/2018] [Indexed: 01/01/2023]
Abstract
Selective serotonin reuptake inhibitor medications (SSRIs) are prescribed to up to 10% of pregnant women to treat maternal mood disorders. Exposure to these medications in-utero has raised concerns about altered neurobehavioral outcomes; most recently those related to peer-to-peer social interactions and play. While clinical data show that both perinatal SSRIs (pSSRI) and maternal stress can contribute to social behavioral changes in children, minimal animal work has investigated the effects of pSSRIs in relevant models of maternal stress or the long-term implications of these effects. Therefore the aim of this work was to investigate the long-term effects of pSSRI exposure to fluoxetine on social behaviors, the hypothalamic pituitary adrenal system (HPA) and hippocampal plasticity in adult male and female rat offspring using a model of pre-gestational maternal stress. Adult Sprague-Dawley female and male rat offspring from the following four groups were utilized: 1. Control + Vehicle, 2. Control + Fluoxetine, 3. Pre-gestational Stress + Vehicle, 4. Pre-gestational Stress + Fluoxetine (n = 8-16/female/age groups, n = 8-14/male/age groups). Main findings show pSSRIs increased social investigation in adult females and increased social play (pouncing, nape attacks) in adult males. Perinatal SSRIs also had sexually differentiated effects on hippocampal neurogenesis and GR density. Pre-gestational stress had enduring effects by decreasing social investigation and hippocampal neurogenesis in adult males. Thus pSSRIs, as well as pre-gestational maternal stress, have significant long-term effects on social neurobehavioral outcomes which differ in males and females. This suggests that it would be valuable to consider fetal-sex specific treatments for maternal mental illness.
Collapse
Affiliation(s)
- Mary Gemmel
- Department of Biological Sciences, Ohio University, Athens, OH, USA
| | | | - Sophia C Mort
- Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - Lesley A Hill
- Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Thierry D Charlier
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Jodi L Pawluski
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|