1
|
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
|
2
|
Puri TA, Lieblich SE, Ibrahim M, Galea LAM. Pregnancy history and estradiol influence spatial memory, hippocampal plasticity, and inflammation in middle-aged rats. Horm Behav 2024; 165:105616. [PMID: 39168073 DOI: 10.1016/j.yhbeh.2024.105616] [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: 05/16/2024] [Revised: 07/28/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Pregnancy and motherhood can have long-term effects on cognition and brain aging in both humans and rodents. Estrogens are related to cognitive function and neuroplasticity. Estrogens can improve cognition in postmenopausal women, but the evidence is mixed, partly due to differences in age of initiation, type of menopause, dose, formulation and route of administration. Additionally, past pregnancy influences brain aging and cognition as a younger age of first pregnancy in humans is associated with poorer aging outcomes. However, few animal studies have examined specific features of pregnancy history or the possible mechanisms underlying these changes. We examined whether maternal age at first pregnancy and estradiol differentially affected hippocampal neuroplasticity, inflammation, spatial reference cognition, and immediate early gene activation in response to spatial memory retrieval in middle-age. Thirteen-month-old rats (who were nulliparous (never mothered) or previously primiparous (had a litter) at three or seven months) received daily injections of estradiol (or vehicle) for sixteen days and were tested on the Morris Water Maze. An older age of first pregnancy was associated with impaired spatial memory but improved performance on reversal training, and increased number of new neurons in the ventral hippocampus. Estradiol decreased activation of new neurons in the dorsal hippocampus, regardless of parity history. Estradiol also decreased the production of anti-inflammatory cytokines based on age of first pregnancy. This work suggests that estradiol affects neuroplasticity and neuroinflammation in middle age, and that age of first pregnancy can have long lasting effects on hippocampus structure and function.
Collapse
Affiliation(s)
- Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Muna Ibrahim
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada; Center for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Lee BH, Cevizci M, Lieblich SE, Ibrahim M, Wen Y, Eid RS, Lamers Y, Duarte-Guterman P, Galea LAM. Exploring the parity paradox: Differential effects on neuroplasticity and inflammation by APOEe4 genotype at middle age. Brain Behav Immun 2024; 120:54-70. [PMID: 38772427 DOI: 10.1016/j.bbi.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 04/20/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024] Open
Abstract
Female sex and Apolipoprotein E (APOE) ε4 genotype are top non-modifiable risk factors for Alzheimer's disease (AD). Although female-unique experiences like parity (pregnancy and motherhood) have positive effects on neuroplasticity at middle age, previous pregnancy may also contribute to AD risk. To explore these seemingly paradoxical long-term effects of parity, we investigated the impact of parity with APOEε4 genotype by examining behavioural and neural biomarkers of brain health in middle-aged female rats. Our findings show that primiparous (parous one time) hAPOEε4 rats display increased use of a non-spatial cognitive strategy and exhibit decreased number and recruitment of new-born neurons in the ventral dentate gyrus of the hippocampus in response to spatial working memory retrieval. Furthermore, primiparity and hAPOEε4 genotype synergistically modulate inflammatory markers in the ventral hippocampus. Collectively, these findings demonstrate that previous parity in hAPOEε4 rats confers an added risk to present with reduced activity and engagement of the hippocampus as well as elevated pro-inflammatory signaling, and underscore the importance of considering female-specific factors and genotype in health research.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Melike Cevizci
- 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
| | - Muna Ibrahim
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Yanhua Wen
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Rand S Eid
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Yvonne Lamers
- Food Nutrition and Health Program, Faculty of Land and Food Systems, University of British Columbia, Vancouver, BC, Canada
| | - 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
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, ON, Canada.
| |
Collapse
|
4
|
Lymer J, Bergman H, Yang S, Mallick R, Galea LAM, Choleris E, Fergusson D. The effects of estrogens on spatial learning and memory in female rodents - A systematic review and meta-analysis. Horm Behav 2024; 164:105598. [PMID: 38968677 DOI: 10.1016/j.yhbeh.2024.105598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/01/2024] [Accepted: 06/19/2024] [Indexed: 07/07/2024]
Abstract
Estrogens have inconsistent effects on learning and memory in both the clinical and preclinical literature. Preclinical literature has the advantage of investigating an array of potentially important factors contributing to the varied effects of estrogens on learning and memory, with stringently controlled studies. This study set out to identify specific factors in the animal literature that influence the effects of estrogens on cognition, for possible translation back to clinical practice. The literature was screened and studies meeting strict inclusion criteria were included in the analysis. Eligible studies included female ovariectomized rodents with an adequate vehicle for the estrogen treatment, with an outcome of spatial learning and memory in the Morris water maze. Training days of the Morris water maze were used to assess acquisition of spatial learning, and the probe trial was used to evaluate spatial memory recall. Continuous outcomes were pooled using a random effects inverse variance method and reported as standardized mean differences with 95 % confidence intervals. Subgroup analyses were developed a priori to assess important factors. The overall analysis favoured treatment for the later stages of training and for the probe trial. Factors including the type of estrogen, route, schedule of administration, age of animals, timing relative to ovariectomy, and duration of treatment were all found to be important. The subgroup analyses showed that chronic treatment with 17β-estradiol, either cyclically or continuously, to young animals improved spatial recall. These results, observed in animals, can inform and guide further clinical research on hormone replacement therapy for cognitive benefits.
Collapse
Affiliation(s)
- Jennifer Lymer
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada.
| | - Hailey Bergman
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | - Sabrina Yang
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | | | - Liisa A M Galea
- Department of Psychiatry, University of Toronto, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada.
| | - Dean Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
5
|
Duarte-Guterman P, Richard JE, Lieblich SE, Eid RS, Lamers Y, Galea LAM. Cellular and molecular signatures of motherhood in the adult and ageing rat brain. Open Biol 2023; 13:230217. [PMID: 37989220 PMCID: PMC10681025 DOI: 10.1098/rsob.230217] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/23/2023] [Indexed: 11/23/2023] Open
Abstract
Pregnancy is marked by robust changes, including brain changes to volume, structure, connectivity and neuroplasticity. Although some brain changes are restricted to pregnancy and the postpartum, others are long-lasting. Few studies have examined possible mechanisms of these changes or the effects of multiple pregnancies. We characterized various cellular and molecular signatures of parity (nulliparous, primiparous, biparous) in the rat hippocampus. We investigated density of neural stems cells (Sox2), microglia (Iba-1) and levels of a synaptic protein (PSD-95), cell signalling pathways, neuroinflammation, and the tryptophan-kynurenine (TRP-KYN) pathway, one week after weaning their pups from the last pregnancy (age of dam: seven months) and in middle-age (age of dam: 13 months). Parity increased PSD-95 levels in both age groups and prevented the age-related decrease in neural stem cell density observed in nulliparous rats. Biparity increased cell signalling phosphoproteins (pp70S6K, S6RP) and number of microglia in the dentate gyrus, regardless of age. Parity resulted in transient changes to the TRP-KYN system. Thus, previous parity has lasting effects on synaptic plasticity with fewer lasting effects on inflammation and cell signalling phosphoproteins in the whole hippocampus.
Collapse
Affiliation(s)
- P. Duarte-Guterman
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain health, University of British Columbia, Vancouver, British Columbia, Canada
| | - J. E. Richard
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain health, University of British Columbia, Vancouver, British Columbia, Canada
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden
| | - S. E. Lieblich
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain health, University of British Columbia, Vancouver, British Columbia, Canada
| | - R. S. Eid
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Y. Lamers
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- 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
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden
| | - L. A. M. Galea
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- 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
| |
Collapse
|
6
|
Koebele SV, Bernaud VE, Northup-Smith SN, Willeman MN, Strouse IM, Bulen HL, Schrier AR, Newbern JM, DeNardo DF, Mayer LP, Dyer CA, Bimonte-Nelson HA. Gynecological surgery in adulthood imparts cognitive and brain changes in rats: A focus on hysterectomy at short-, moderate-, and long-term intervals after surgery. Horm Behav 2023; 155:105411. [PMID: 37659358 PMCID: PMC11060888 DOI: 10.1016/j.yhbeh.2023.105411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/11/2023] [Accepted: 08/08/2023] [Indexed: 09/04/2023]
Abstract
Premenopausal hysterectomy is associated with a greater relative risk of dementia. We previously demonstrated cognitive impairments in adult rats six weeks after hysterectomy with ovarian conservation compared with intact sham-controls and other gynecological surgery variations. Here, we investigated whether hysterectomy-induced cognitive impairments are transient or persistent. Adult rats received sham-control, ovariectomy (Ovx), hysterectomy, or Ovx-hysterectomy surgery. Spatial working memory, reference memory, and anxiety-like behavior were tested either six-weeks post-surgery, in adulthood; seven-months post-surgery, in early middle-age; or twelve-months post-surgery, in late middle-age. Hysterectomy in adulthood yielded spatial working memory deficits at short-, moderate-, and long-term post-surgery intervals. Serum hormone levels did not differ between ovary-intact, but differed from Ovx, groups. Hysterectomy had no significant impact on healthy ovarian follicle or corpora lutea counts for any post-surgery timepoint compared with intact sham-controls. Frontal cortex, dorsal hippocampus, and entorhinal cortex were assessed for activity-dependent markers. In entorhinal cortex, there were alterations in FOSB and ΔFOSB expression during the early middle-age timepoint, and phosphorylated ERK1/2 levels at the adult timepoint. Collectively, results suggest a primary role for the uterus in regulating cognition, and that memory-related neural pathways may be modified following gynecological surgery. This is the first preclinical report of long-term effects of hysterectomy with and without ovarian conservation on cognition, endocrine, ovarian, and brain assessments, initiating a comprehensive framework of gynecological surgery effects. Translationally, findings underscore critical needs to decipher how gynecological surgeries, especially those involving the uterus, impact the brain and its functions, the ovaries, and overall aging from a systems perspective.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Victoria E Bernaud
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Steven N Northup-Smith
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Mari N Willeman
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Isabel M Strouse
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Haidyn L Bulen
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Ally R Schrier
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Dale F DeNardo
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | | | | | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA.
| |
Collapse
|
7
|
Puri TA, Richard JE, Galea LAM. Beyond sex differences: short- and long-term effects of pregnancy on the brain. Trends Neurosci 2023; 46:459-471. [PMID: 37120339 DOI: 10.1016/j.tins.2023.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 05/01/2023]
Abstract
Growing attention has been directed to the inclusion of females in neuroscience studies, and to the importance of studying sex as a biological variable. However, how female-specific factors such as menopause and pregnancy, affect the brain remains understudied. In this review, we use pregnancy as a case in point of a female-unique experience that can alter neuroplasticity, neuroinflammation, and cognition. We examine studies in both humans and rodents indicating that pregnancy can modify neural function in the short term, as well as alter the trajectory of brain aging. Furthermore, we discuss the influence of maternal age, fetal sex, number of pregnancies, and presence of pregnancy complications on brain health outcomes. We conclude by encouraging the scientific community to prioritize researching female health by recognizing and including factors such as pregnancy history in research.
Collapse
Affiliation(s)
- Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Jennifer E Richard
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada; Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
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
|
9
|
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: 2.3] [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
|
10
|
Been LE, Sheppard PAS, Galea LAM, Glasper ER. Hormones and neuroplasticity: A lifetime of adaptive responses. Neurosci Biobehav Rev 2021; 132:679-690. [PMID: 34808191 DOI: 10.1016/j.neubiorev.2021.11.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022]
Abstract
Major life transitions often co-occur with significant fluctuations in hormones that modulate the central nervous system. These hormones enact neuroplastic mechanisms that prepare an organism to respond to novel environmental conditions and/or previously unencountered cognitive, emotional, and/or behavioral demands. In this review, we will explore several examples of how hormones mediate neuroplastic changes in order to produce adaptive responses, particularly during transitions in life stages. First, we will explore hormonal influences on social recognition in both males and females as they transition to sexual maturity. Next, we will probe the role of hormones in mediating the transitions to motherhood and fatherhood, respectively. Finally, we will survey the long-term impact of reproductive experience on neuroplasticity in females, including potential protective effects and risk factors associated with reproductive experience in mid-life and beyond. Ultimately, a more complete understanding of how hormones influence neuroplasticity throughout the lifespan, beyond development, is necessary for understanding how individuals respond to life changes in adaptive ways.
Collapse
Affiliation(s)
- Laura E Been
- Department of Psychology, Haverford College, 370 Lancaster Avenue, Haverford, PA, 19041, USA.
| | - Paul A S Sheppard
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, 1151 Richmond St, London, Ontario, N6A 5B7, Canada.
| | - Liisa A M Galea
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada.
| | - Erica R Glasper
- Department of Psychology, University of Maryland, College Park, MD, 20742 USA.
| |
Collapse
|
11
|
Medina J, De Guzman RM, Workman JL. Lactation is not required for maintaining maternal care and active coping responses in chronically stressed postpartum rats: Interactions between nursing demand and chronic variable stress. Horm Behav 2021; 136:105035. [PMID: 34488064 DOI: 10.1016/j.yhbeh.2021.105035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/11/2021] [Accepted: 07/04/2021] [Indexed: 10/20/2022]
Abstract
Women who do not breastfeed or discontinue breastfeeding early are more likely to develop postpartum depression (PPD) and stress is a significant risk factor for depression, including PPD. Using a rat model, we investigated whether the absence of nursing would increase the susceptibility to chronic stress-related behavioral and neural changes during the postpartum period. Adult female rats underwent thelectomy (thel; removal of teats), sham surgery, or no surgery (control) and were paired with males for breeding. All litters were rotated twice daily until postpartum day (PD) 26. Sham rats served as surrogates for thel litters, yielding a higher nursing demand for sham rats. Concurrently, rats received either no stress or chronic variable stress until PD 25. Rats were observed for maternal behaviors and tested in a series of tasks including open field, sucrose preference, and forced swim. We used immunohistochemistry (IHC) for doublecortin (DCX; to label immature neurons) or for mineralocorticoid receptor (MR). Contrary to our expectations, non-nursing thel rats were resistant to the effects of stress in all dependent measures. Our data indicate that even in chronic adverse conditions, nursing is not required for maintaining stable care to offspring or active coping responses in an acutely stressful task. We discuss the possible role of offspring contact and consider future directions for biomedical and clinical research. In rats with high nursing demand, however, chronic stress increased immobility, hippocampal neurogenesis, and MR expression (largely in opposition to the effects of stress in rats with typical nursing demand). We discuss these patterns in the context of energetics and allostatic load. This research highlights the complexity in relationships between stress, nursing, and neurobehavioral outcomes in the postpartum period and underscores the need for additional biomedical and clinical research geared toward optimizing treatments and interventions for women with PPD, regardless of breastfeeding status. SIGNIFICANCE STATEMENT: The goal of this research was to determine how the absence of nursing and higher nursing demand impact stress-coping behaviors and neural changes associated with chronic stress in order to disentangle the complex interplay of factors that contribute to psychological illness during the postpartum period.
Collapse
Affiliation(s)
- Joanna Medina
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America
| | - Rose M De Guzman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America
| | - Joanna L Workman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America; Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Ave., Albany, NY 12222, United States of America.
| |
Collapse
|
12
|
Mouihate A, Kalakh S. Maternal Interleukin-6 Hampers Hippocampal Neurogenesis in Adult Rat Offspring in a Sex-Dependent Manner. Dev Neurosci 2021; 43:106-115. [PMID: 34023825 DOI: 10.1159/000516370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/19/2021] [Indexed: 11/19/2022] Open
Abstract
Maternal immune activation (MIA) during pregnancy leads to long-lasting effects on brain development and function. Several lines of evidence suggest that the maternal inflammatory cytokine interleukin (IL)-6 plays a crucial role in the long-lasting effects of MIA on adult offspring. IL-6 is naturally produced during pregnancy in the absence of any underlying immune activation. The objective of this study was to assess whether this naturally occurring IL-6 has long-lasting effects on brain plasticity and function. Therefore, pregnant rats were given either an IL-6-neutralizing antibody (IL-6Ab) or vehicle during the third week of pregnancy. Newly born (doublecortin) and mature neurons (NeuN) were monitored in the hippocampus of adult male and female offspring. Prenatal IL-6Ab led to an enhanced number of newly born and mature neurons in the dentate gyrus of the hippocampus of male but not female adult offspring. This enhanced neurogenesis was associated with an increased propensity in memory acquisition in male offspring. Blunting the naturally occurring IL-6 during pregnancy did not have a significant long-lasting impact on astrocyte cell density (GFAP), or on anxiety-like behavior as assessed with elevated plus maze and open field tests. Taken together, these data suggest that maternal IL-6 contributes, at least in part, to the programming of the brain's development in a sex-dependent manner.
Collapse
Affiliation(s)
- Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Samah Kalakh
- Department of Physiology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| |
Collapse
|
13
|
Ladyman SR, Brooks VL. Central actions of insulin during pregnancy and lactation. J Neuroendocrinol 2021; 33:e12946. [PMID: 33710714 PMCID: PMC9198112 DOI: 10.1111/jne.12946] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/24/2021] [Accepted: 01/27/2021] [Indexed: 12/17/2022]
Abstract
Pregnancy and lactation are highly metabolically demanding states. Maternal glucose is a key fuel source for the growth and development of the fetus, as well as for the production of milk during lactation. Hence, the maternal body undergoes major adaptations in the systems regulating glucose homeostasis to cope with the increased demand for glucose. As part of these changes, insulin levels are elevated during pregnancy and lower in lactation. The increased insulin secretion during pregnancy plays a vital role in the periphery; however, the potential effects of increased insulin action in the brain have not been widely investigated. In this review, we consider the impact of pregnancy on brain access and brain levels of insulin. Moreover, we explore the hypothesis that pregnancy is associated with site-specific central insulin resistance that is adaptive, allowing for the increases in peripheral insulin secretion without the consequences of increased central and peripheral insulin functions, such as to stimulate glucose uptake into maternal tissues or to inhibit food intake. Conversely, the loss of central insulin actions may impair other functions, such as insulin control of the autonomic nervous system. The potential role of low insulin in facilitating adaptive responses to lactation, such as hyperphagia and suppression of reproductive function, are also discussed. We end the review with a list of key research questions requiring resolution.
Collapse
Affiliation(s)
- Sharon R Ladyman
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Virginia L Brooks
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
14
|
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: 0.5] [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
|
15
|
Achiron A, Ben-David A, Gurevich M, Magalashvili D, Menascu S, Dolev M, Stern Y, Ziv-Baran T. Parity and disability progression in relapsing-remitting multiple sclerosis. J Neurol 2020; 267:3753-3762. [PMID: 32725314 DOI: 10.1007/s00415-020-10093-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 10/23/2022]
Abstract
AIM It is unclear whether parity and increasing parity are risk factors for long-term disability progression in relapsing-remitting multiple sclerosis. Furthermore, data on the effects of immunomodulatory treatments in this context are limited. OBJECTIVES To examine the association between parity and long-term neurological sequela among relapsing-remitting multiple sclerosis patients. METHODS A cohort study including all women with relapsing-remitting multiple sclerosis in Israel registered in Sheba Medical Center Multiple Sclerosis data registry from 1995 to 2018. The risks of progression to moderate and severe disability according to parity after disease onset were evaluated. Cox regression models using childbirth as a time-dependent covariate were used to study the association between parity and disability progression. RESULTS During the 26,785 person-years of follow-up a total of 2281 women were included in the study. Parity was associated with decreased risk of progression to moderate (adj.HR, 0.68; 95% CI 0.54-0.85, P = 0.001) but not to severe disability (adj.HR, 0.88; 95% CI 0.68-1.14, P = 0.36). Hazard ratios for progression to moderate and severe disability were comparable between women with one, two, and three or more births. In a subgroup analysis of women who gave birth within 5 years of disease onset, immunomodulatory treatment did not affect moderate or severe disability-free survival. CONCLUSION This study suggests that childbirth after the onset of multiple sclerosis is associated with a decreased risk of progression to moderate neurological disability.
Collapse
Affiliation(s)
- Anat Achiron
- Multiple Sclerosis Center, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Alon Ben-David
- Department of Obstetrics and Gynecology, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michael Gurevich
- Multiple Sclerosis Center, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - David Magalashvili
- Multiple Sclerosis Center, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shay Menascu
- Multiple Sclerosis Center, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Mark Dolev
- Multiple Sclerosis Center, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yael Stern
- Multiple Sclerosis Center, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Tomer Ziv-Baran
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
16
|
Barth C, de Lange AMG. Towards an understanding of women's brain aging: the immunology of pregnancy and menopause. Front Neuroendocrinol 2020; 58:100850. [PMID: 32504632 DOI: 10.1016/j.yfrne.2020.100850] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/23/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
Abstract
Women are at significantly greater risk of developing Alzheimer's disease and show higher prevalence of autoimmune conditions relative to men. Women's brain health is historically understudied, and little is therefore known about the mechanisms underlying epidemiological sex differences in neurodegenerative diseases, and how female-specific factors may influence women's brain health across the lifespan. In this review, we summarize recent studies on the immunology of pregnancy and menopause, emphasizing that these major immunoendocrine transition phases may play a critical part in women's brain aging trajectories.
Collapse
Affiliation(s)
- Claudia Barth
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Ann-Marie G de Lange
- Department of Psychology, University of Oslo, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK.
| |
Collapse
|
17
|
Hippocampal Subregion Transcriptomic Profiles Reflect Strategy Selection during Cognitive Aging. J Neurosci 2020; 40:4888-4899. [PMID: 32376783 DOI: 10.1523/jneurosci.2944-19.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/08/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
Age-related cognitive impairments are associated with differentially expressed genes (DEGs) linked to defined neural systems; however, studies examining multiple regions of the hippocampus fail to find links between behavior and transcription in the dentate gyrus (DG). We hypothesized that use of a task requiring intact DG function would emphasize molecular signals in the DG associated with a decline in performance. We used a water maze beacon discrimination task to characterize young and middle-age male F344 rats, followed by a spatial reference memory probe trial test. Middle-age rats showed increased variability in discriminating two identical beacons. Use of an allocentric strategy and formation of a spatial reference memory were not different between age groups; however, older animals compensated for impaired beacon discrimination through greater reliance on spatial reference memory. mRNA sequencing of hippocampal subregions indicated DEGs in the DG of middle-age rats, linked to synaptic function and neurogenesis, correlated with beacon discrimination performance, suggesting that senescence of the DG underlies the impairment. Few genes correlated with spatial memory across age groups, with a greater number in region CA1. Age-related CA1 DEGs, correlated with spatial memory, were linked to regulation of neural activity. These results indicate that the beacon task is sensitive to impairment in middle age, and distinct gene profiles are observed in neural circuits that underlie beacon discrimination performance and allocentric memory. The use of different strategies in older animals and associated transcriptional profiles could provide an animal model for examining cognitive reserve and neural compensation of aging.SIGNIFICANCE STATEMENT Hippocampal subregions are thought to differentially contribute to memory. We took advantage of age-related variability in performance on a water maze beacon task and next-generation sequencing to test the hypothesis that aging of the dentate gyrus is linked to impaired beacon discrimination and compensatory use of allocentric memory. The dentate gyrus expressed synaptic function and neurogenesis genes correlated with beacon discrimination in middle-age animals. Spatial reference memory was associated with CA1 transcriptional correlates linked to regulation of neural activity and use of an allocentric strategy. This is the first study examining transcriptomes of multiple hippocampal subregions to link age-related impairments associated with discrimination of feature overlap and alternate response strategies to gene expression in specific hippocampal subregions.
Collapse
|
18
|
Deems NP, Leuner B. Pregnancy, postpartum and parity: Resilience and vulnerability in brain health and disease. Front Neuroendocrinol 2020; 57:100820. [PMID: 31987814 PMCID: PMC7225072 DOI: 10.1016/j.yfrne.2020.100820] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/25/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Risk and resilience in brain health and disease can be influenced by a variety of factors. While there is a growing appreciation to consider sex as one of these factors, far less attention has been paid to sex-specific variables that may differentially impact females such as pregnancy and reproductive history. In this review, we focus on nervous system disorders which show a female bias and for which there is data from basic research and clinical studies pointing to modification in disease risk and progression during pregnancy, postpartum and/or as a result of parity: multiple sclerosis (MS), depression, stroke, and Alzheimer's disease (AD). In doing so, we join others (Shors, 2016; Galea et al., 2018a) in aiming to illustrate the importance of looking beyond sex in neuroscience research.
Collapse
Affiliation(s)
- Nicholas P Deems
- The Ohio State University, Department of Psychology, Columbus, OH, USA
| | - Benedetta Leuner
- The Ohio State University, Department of Psychology, Columbus, OH, USA.
| |
Collapse
|
19
|
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: 2.5] [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
|
20
|
Harville EW, Guralnik J, Romero M, Bazzano LA. Reproductive History and Cognitive Aging: The Bogalusa Heart Study. Am J Geriatr Psychiatry 2020; 28:217-225. [PMID: 31350162 PMCID: PMC6942641 DOI: 10.1016/j.jagp.2019.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/08/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Although it has become increasingly clear that pregnancy-related health predicts later-life cardiometabolic health, the relationship between reproductive history and cognitive health is less frequently studied. Although some research has identified associations between parity or hypertensive disorders of pregnancy and cognitive changes, the evidence is mixed. OBJECTIVE To examine the association between reproductive history and midlife cognition in a community-based population. STUDY DESIGN Seven hundred and thirty midlife women in the Bogalusa Heart Study completed a brief cognitive battery (memory, attention, executive function, and processing speed) and were interviewed about their reproductive history. Reproductive history (parity, age at first pregnancy, and breastfeeding) and pregnancy complications (low birthweight, preterm birth, hypertensive disorders, and miscarriage) were examined as predictors of cognitive function, with adjustment for potential confounders. RESULTS Nulliparous women had an overall lower cognitive score (adjusted beta -1.50, standard error [SE]: 0.41). Adolescent birth was associated with a somewhat better performance on the Trail Making Test (beta -0.31, SE: 0.15 for birth <16 years), while high parity was not strongly associated with any of the cognitive measures. History of pregnancy complications was not strongly associated with cognitive function, whereas history of miscarriage was associated with better cognitive function, as was a history of breastfeeding (beta overall score 0.90, SE: 0.29), particularly noticeable for semantic memory and in those with more total breastfeeding time (beta for overall score among those with >24 weeks lifetime breastfeeding, beta 1.21, SE: 0.44). CONCLUSION Nulliparity and breastfeeding are associated with midlife cognition in women. Future studies should examine possible mechanisms by which these associations are created.
Collapse
Affiliation(s)
- Emily W Harville
- Department of Epidemiology (EWH, LAB), Tulane School of Public Health and Tropical Medicine, New Orleans, LA.
| | - Jack Guralnik
- Department of Epidemiology and Public Health, School of Medicine (JG), University of Maryland, Baltimore, MD
| | - Maryellen Romero
- Department of Psychiatry and Behavioral Sciences (MR), Tulane University Health Sciences Center, New Orleans, LA
| | - Lydia A Bazzano
- Department of Epidemiology (EWH, LAB), Tulane School of Public Health and Tropical Medicine, New Orleans, LA
| |
Collapse
|
21
|
Population-based neuroimaging reveals traces of childbirth in the maternal brain. Proc Natl Acad Sci U S A 2019; 116:22341-22346. [PMID: 31615888 DOI: 10.1073/pnas.1910666116] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Maternal brain adaptations have been found across pregnancy and postpartum, but little is known about the long-term effects of parity on the maternal brain. Using neuroimaging and machine learning, we investigated structural brain characteristics in 12,021 middle-aged women from the UK Biobank, demonstrating that parous women showed less evidence of brain aging compared to their nulliparous peers. The relationship between childbirths and a "younger-looking" brain could not be explained by common genetic variation or relevant confounders. Although prospective longitudinal studies are needed, the results suggest that parity may involve neural changes that could influence women's brain aging later in life.
Collapse
|
22
|
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: 5.6] [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
|
23
|
Chronic aromatase inhibition increases ventral hippocampal neurogenesis in middle-aged female mice. Psychoneuroendocrinology 2019; 106:111-116. [PMID: 30974324 DOI: 10.1016/j.psyneuen.2019.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/02/2019] [Indexed: 12/21/2022]
Abstract
Letrozole, a third-generation aromatase inhibitor, prevents the production of estrogens in the final step in conversion from androgens. Due to its efficacy at suppressing estrogens, letrozole has recently taken favor as a first-line adjuvant treatment for hormone-responsive breast cancer in middle-aged women. Though patient response to letrozole has generally been positive, there is conflicting evidence surrounding its effects on the development of depression. It is possible that the potential adverse effects of letrozole on mood are a result of the impact of hormonal fluctuations on neurogenesis in the hippocampus. Thus, to clarify the effects of letrozole on the hippocampus and behavior, we examined how chronic administration affects hippocampal neurogenesis and depressive-like behavior in middle-aged, intact female mice. Mice were given either letrozole (1 mg/kg) or vehicle by injection (i.p.) daily for 3 weeks. Depressive-like behavior was assessed during the last 3 days of treatment using the forced swim test, tail suspension test, and sucrose preference test. The production of new neurons was quantified using the immature neuronal marker doublecortin (DCX), and cell proliferation was quantified using the endogenous marker Ki67. We found that letrozole increased DCX and Ki67 expression and maturation in the dentate gyrus, but had no significant effect on depressive-like behavior. Our findings suggest that a reduction in estrogens in middle-aged females increases hippocampal neurogenesis without any adverse impact on depressive-like behavior; as such, this furthers our understanding of how estrogens modulate neurogenesis, and to the rationale for the utilization of letrozole in the clinical management of breast cancer.
Collapse
|
24
|
Rogers FD, Bales KL. Mothers, Fathers, and Others: Neural Substrates of Parental Care. Trends Neurosci 2019; 42:552-562. [PMID: 31255381 PMCID: PMC6660995 DOI: 10.1016/j.tins.2019.05.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/29/2019] [Accepted: 05/23/2019] [Indexed: 12/29/2022]
Abstract
Parental care is essential for the survival of offspring in altricial mammalian species. However, in most mammals, virgin females tend to avoid or attack infants. Moreover, most males demonstrate avoidance and aggression toward infants, and have little to no involvement in parental care. What mechanisms suppress avoidance, and support approach towards pups, to promote maternal care? In biparental and cooperatively breeding species, what mechanisms allow nonmothers (i.e., fathers and alloparents) to demonstrate parental care? In this review we consider the mechanisms that subserve parental care in mothers, fathers, and others (i.e., alloparents). We emphasize recent discoveries and research trends with particular emphasis on neuroendocrinology, neuroplasticity, transcriptomics, and epigenetics. Finally, we consider outstanding questions and outline opportunities for future research.
Collapse
Affiliation(s)
- Forrest Dylan Rogers
- Graduate Program in Psychology, University of California, Davis, CA 95616, USA; Department of Psychology, University of California, Davis, CA 95616, USA
| | - Karen Lisa Bales
- Department of Psychology, University of California, Davis, CA 95616, USA; California National Primate Research Center, Davis, CA 95616, USA.
| |
Collapse
|
25
|
Taylor CM, Pritschet L, Yu S, Jacobs EG. Applying a Women's Health Lens to the Study of the Aging Brain. Front Hum Neurosci 2019; 13:224. [PMID: 31333434 PMCID: PMC6625223 DOI: 10.3389/fnhum.2019.00224] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/18/2019] [Indexed: 11/15/2022] Open
Abstract
A major challenge in neuroscience is to understand what happens to a brain as it ages. Such insights could make it possible to distinguish between individuals who will undergo typical aging and those at risk for neurodegenerative disease. Over the last quarter century, thousands of human brain imaging studies have probed the neural basis of age-related cognitive decline. "Aging" studies generally enroll adults over the age of 65, a historical precedent rooted in the average age of retirement. A consequence of this research tradition is that it overlooks one of the most significant neuroendocrine changes in a woman's life: the transition to menopause. The menopausal transition is marked by an overall decline in ovarian sex steroid production-up to 90% in the case of estradiol-a dramatic endocrine change that impacts multiple biological systems, including the brain. Despite sex differences in the risk for dementia, the influence that biological sex and sex hormones have on the aging brain is historically understudied, leaving a critical gap in our understanding of the aging process. In this Perspective article, we highlight the influence that endocrine factors have on the aging brain. We devote particular attention to the neural and cognitive changes that unfold in the middle decade of life, as a function of reproductive aging. We then consider emerging evidence from animal and human studies that other endocrine factors occurring earlier in life (e.g., pregnancy, hormonal birth control use) also shape the aging process. Applying a women's health lens to the study of the aging brain will advance knowledge of the neuroendocrine basis of cognitive aging and ensure that men and women get the full benefit of our research efforts.
Collapse
Affiliation(s)
- Caitlin M. Taylor
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
- The Sage Center for the Study of the Mind, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Laura Pritschet
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Shuying Yu
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Emily G. Jacobs
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
26
|
Eid RS, Chaiton JA, Lieblich SE, Bodnar TS, Weinberg J, Galea LAM. Early and late effects of maternal experience on hippocampal neurogenesis, microglia, and the circulating cytokine milieu. Neurobiol Aging 2019; 78:1-17. [PMID: 30825663 DOI: 10.1016/j.neurobiolaging.2019.01.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/03/2019] [Accepted: 01/21/2019] [Indexed: 01/13/2023]
Abstract
The maternal brain displays considerable plasticity, and motherhood is associated with changes in affective and cognitive function. Motherhood can alter the trajectory of brain aging, including modifications to neuroplasticity and cognition. Here, we investigated the short- and long-term effects of motherhood on hippocampal neurogenesis, microglial density and morphology, and circulating cytokines, domains known to be altered with age and implicated in cognition and mood. Female rats were bred then euthanized during gestation or at various postpartum time points, culminating in middle age, and nulliparous rats served as age-matched controls. Hippocampal neurogenesis was significantly suppressed during gestation and the postpartum period. Interestingly, neurogenesis declined significantly in middle-aged nulliparous rats but increased in primiparous rats across the same period. Transient postpartum adaptations to the neuroimmune environment of the hippocampus were evidenced, as Iba-1-immunoreactive microglia assumed a deramified morphology followed by increased density. Intriguingly, aging-related changes in circulating cytokines were dependent on parity. These adaptations in neurogenic and immune processes may have ramifications for maternal mood and cognition across the peripartum period and beyond.
Collapse
Affiliation(s)
- Rand S Eid
- 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
| | - Jessica A Chaiton
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada; Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada; Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tamara S Bodnar
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joanne Weinberg
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, 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; Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
27
|
Pregnancy Promotes Maternal Hippocampal Neurogenesis in Guinea Pigs. Neural Plast 2019; 2019:5765284. [PMID: 31097956 PMCID: PMC6487096 DOI: 10.1155/2019/5765284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/08/2019] [Accepted: 02/21/2019] [Indexed: 11/26/2022] Open
Abstract
Adult neurogenesis in the hippocampal dentate gyrus (DG) modulates cognition and behavior in mammals, while motherhood is associated with cognitive and behavioral changes essential for the care of the young. In mice and rats, hippocampal neurogenesis is reported to be reduced or unchanged during pregnancy, with few data available from other species. In guinea pigs, pregnancy lasts ~9 weeks; we set to explore if hippocampal neurogenesis is altered in these animals, relative to gestational stages. Time-pregnant primigravidas (3-5 months old) and age-matched nonpregnant females were examined, with neurogenic potential evaluated via immunolabeling of Ki67, Sp8, doublecortin (DCX), and neuron-specific nuclear antigen (NeuN) combined with bromodeoxyuridine (BrdU) birth-dating. Relative to control, subgranular Ki67, Sp8, and DCX-immunoreactive (+) cells tended to increase from early gestation to postpartum and peaked at the late gestational stage. In BrdU pulse-chasing experiments in nonpregnant females surviving for different time points (2-120 days), BrdU+ cells in the DG colocalized with DCX partially from 2 to 42 days (most frequently at 14-30 days) and with NeuN increasingly from 14 to 120 days. In pregnant females that received BrdU at early, middle, and late gestational stages and survived for 42 days, the density of BrdU+ cells in the DG was mostly high in the late gestational group. The rates of BrdU/DCX and BrdU/NeuN colocalization were similar among these groups and comparable to those among the corresponding control group. Together, the findings suggest that pregnancy promotes maternal hippocampal neurogenesis in guinea pigs, at least among primigravidas.
Collapse
|
28
|
Duarte-Guterman P, Leuner B, Galea LAM. The long and short term effects of motherhood on the brain. Front Neuroendocrinol 2019; 53:100740. [PMID: 30826374 DOI: 10.1016/j.yfrne.2019.02.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/25/2019] [Accepted: 02/21/2019] [Indexed: 12/13/2022]
Abstract
Becoming a mother is associated with dramatic changes in physiology, endocrinology, immune function, and behaviour that begins during pregnancy and persists into the postpartum. Evidence also suggests that motherhood is accompanied by long-term changes in brain function. In this review, we summarize the short (pregnancy and postpartum) and long-term (beyond the postpartum and into middle age) effects of pregnancy and motherhood on cognition, neuroplasticity, and neuroimmune signalling. We also discuss the effects of previous history of pregnancy and motherhood (parity) on brain health and disease (neurodegenerative diseases and stroke outcomes) and on efficacy of hormone and antidepressant treatments. Finally, we argue that pregnancy and motherhood are unique female experiences that need to be taken into account to better understand female brain function and aging.
Collapse
Affiliation(s)
- Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, The University of British Columbia, Vancouver, BC, Canada
| | - Benedetta Leuner
- Department of Psychology and Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, The University of British Columbia, Vancouver, BC, Canada. http://galealab.psych.ubc.ca
| |
Collapse
|
29
|
Waters EM, Mazid S, Dodos M, Puri R, Janssen WG, Morrison JH, McEwen BS, Milner TA. Effects of estrogen and aging on synaptic morphology and distribution of phosphorylated Tyr1472 NR2B in the female rat hippocampus. Neurobiol Aging 2019; 73:200-210. [PMID: 30384123 PMCID: PMC11548941 DOI: 10.1016/j.neurobiolaging.2018.09.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/10/2018] [Accepted: 09/18/2018] [Indexed: 12/12/2022]
Abstract
Age and estrogens may impact the mobility of N-methyl-D-aspartate receptors (NMDARs) in hippocampal synapses. Here, we used serial section immunogold electron microscopy to examine whether phosphorylated tyrosine 1472 NR2B (pY1472), which is involved in the surface expression of NMDARs, is altered in the dorsal hippocampus of young (3-4 months old) and aged (∼24 months old) ovariectomized rats treated with 17β-estradiol or vehicle for 2 days. The number of gold particles labeling pY1472 was higher in presynaptic and postsynaptic compartments of aged rats with low estradiol (vehicle-treated) compared to other groups. In terminals, pY1472 levels were elevated in aged rats but reduced by estradiol treatment to levels seen in young rats. Conversely, the mitochondria number was lower in aged females but was restored to young levels by estradiol. In the postsynaptic density and dendritic spines, estradiol reduced pY1472 in young and aged rats. As phosphorylation at Y1472 blocks NR2B endocytosis, reduction of pY1472 by estradiol suggests another mechanism through which estrogen enhances synaptic plasticity by altering localization of NMDAR subunits within synapses.
Collapse
Affiliation(s)
- Elizabeth M Waters
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Sanoara Mazid
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Mariana Dodos
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Rishi Puri
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William G Janssen
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John H Morrison
- Department of Neurology, Center for Neuroscience, The California National Primate Research Center, UC Davis, Davis, CA, USA
| | - Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Teresa A Milner
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
30
|
Beyond sex differences: short and long-term implications of motherhood on women’s health. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
31
|
Galea LA, Roes MM, Dimech CJ, Chow C, Mahmoud R, Lieblich SE, Duarte-Guterman P. Premarin has opposing effects on spatial learning, neural activation, and serum cytokine levels in middle-aged female rats depending on reproductive history. Neurobiol Aging 2018; 70:291-307. [DOI: 10.1016/j.neurobiolaging.2018.06.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 06/23/2018] [Accepted: 06/24/2018] [Indexed: 01/28/2023]
|
32
|
Medina J, Workman JL. Maternal experience and adult neurogenesis in mammals: Implications for maternal care, cognition, and mental health. J Neurosci Res 2018; 98:1293-1308. [DOI: 10.1002/jnr.24311] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 07/07/2018] [Accepted: 07/11/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Joanna Medina
- Department of Psychology and the Center for Neuroscience Research University at Albany, State University of New York New York
| | - Joanna L. Workman
- Department of Psychology and the Center for Neuroscience Research University at Albany, State University of New York New York
| |
Collapse
|
33
|
Korol DL, Wang W. Using a memory systems lens to view the effects of estrogens on cognition: Implications for human health. Physiol Behav 2018; 187:67-78. [PMID: 29203121 PMCID: PMC5844822 DOI: 10.1016/j.physbeh.2017.11.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 01/23/2023]
Abstract
Understanding the organizing and activating effects of gonadal steroids on adult physiology can guide insight into sex differences in and hormonal influences on health and disease, ranging from diabetes and other metabolic disorders, emotion and stress regulation, substance abuse, pain perception, immune function and inflammation, to cognitive function and dysfunction accompanying neurological disorders. Because the brain is highly sensitive to many forms of estrogens, it is not surprising that many adult behaviors, including cognitive function, are modulated by estrogens. Estrogens are known for their facilitating effects on learning and memory, but it is becoming increasingly clear that they also can impair learning and memory of some classes of tasks and may do so through direct actions on specific neural systems. This review takes a multiple memory systems approach to understanding how estrogens can at the same time enhance hippocampus-sensitive place learning and impair striatum-sensitive response learning by exploring the role estrogen receptor signaling may play in the opposing cognitive effects of estrogens. Accumulating evidence suggests that neither receptor subtype nor the timing of treatment, i.e. rapid vs slow, explain the bidirectional effects of estrogens on different types of learning. New findings pointing to neural metabolism and the provision of energy substrates by astrocytes as a candidate mechanism for cognitive enhancement and impairment are discussed.
Collapse
Affiliation(s)
- Donna L Korol
- Department of Biology, Syracuse University, Syracuse, NY 13244, United States.
| | - Wei Wang
- Department of Biology, Syracuse University, Syracuse, NY 13244, United States
| |
Collapse
|
34
|
Gheorghe A, Qiu W, Galea LAM. Hormonal Regulation of Hippocampal Neurogenesis: Implications for Depression and Exercise. Curr Top Behav Neurosci 2018; 43:379-421. [PMID: 30414016 DOI: 10.1007/7854_2018_62] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adult hippocampal neurogenesis exists in all mammalian species, including humans, and although there has been considerable research investigating the function and regulation of neurogenesis, there remain many open questions surrounding the complexity of this phenomenon. This stems partially from the fact that neurogenesis is a multistage process that involves proliferation, differentiation, migration, survival, and eventual integration of new cells into the existing hippocampal circuitry, each of which can be independently influenced. The function of adult neurogenesis in the hippocampus is related to stress regulation, behavioral efficacy of antidepressants, long-term spatial memory, forgetting, and pattern separation. Steroid hormones influence the regulation of hippocampal neurogenesis, stress regulation, and cognition and differently in males and females. In this chapter, we will briefly tap into the complex network of steroid hormone modulation of neurogenesis in the hippocampus with specific emphasis on stress, testosterone, and estrogen. We examine the possible role of neurogenesis in the etiology of depression and influencing treatment by examining the influence of both pharmacological (selective serotonin reuptake inhibitors, tricyclic antidepressants) treatments and non-pharmacological (exercise) remedies.
Collapse
Affiliation(s)
- Ana Gheorghe
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada. .,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada. .,Department of Psychology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
35
|
He Q, Luo Y, Lv F, Xiao Q, Chao F, Qiu X, Zhang L, Gao Y, Xiu Y, Huang C, Tang Y. Effects of estrogen replacement therapy on the myelin sheath ultrastructure of myelinated fibers in the white matter of middle-aged ovariectomized rats. J Comp Neurol 2017; 526:790-802. [PMID: 29205359 DOI: 10.1002/cne.24366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/17/2022]
Abstract
The effects of estrogen replacement therapy (ORT) on white matter and the myelin sheath ultrastructure in the white matter of middle-aged ovariectomized (OVX) rats were investigated in this study. Middle-aged rats were ovariectomized and divided into a placebo replacement (OVX + O) group and an estrogen replacement (OVX + E) group. Then, the Morris water maze, electron microscope techniques, and stereological methods were used to investigate the effects of ORT on spatial learning capacity, white matter volume and the myelin sheath ultrastructure in the white matter. We found that the spatial learning capacity of the OVX + E rats was significantly improved compared with that of the OVX + O rats. When compared with that of OVX + O rats, the total volume of the myelin sheaths in the white matter of the OVX + E rats was significantly increased by 27%, and the difference between the outer perimeter and inner perimeter of the myelin sheaths of the white matter in the OVX + E rats increased significantly by 12.6%. The myelinated fibers with mean diameters of 1.2-1.4 μm were significantly longer (46.1%) in the OVX + E rats; the difference between the mean diameter of myelinated fibers and the mean diameter of axons (0-0.4 μm) was significantly increased by 21.6% in the OVX + E rats. These results suggested that ORT had positive protective effects on the spatial learning ability and on the myelin sheath ultrastructure in the white matter of middle-aged OVX rats.
Collapse
Affiliation(s)
- Qi He
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yanmin Luo
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Fulin Lv
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Qian Xiao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Fenglei Chao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Xuan Qiu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China.,Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Ministry of Education, Chongqing Medical University, Chongqing, P. R. China
| | - Lei Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yuan Gao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China.,Department of Geriatrics, The First Affiliated Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Yun Xiu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Institute of Life Science, Chongqing Medical University, Chongqing, P. R. China
| | - Chunxia Huang
- Department of Physiology, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
36
|
Abstract
The brain has long been known as a dimorphic organ and as a target of sex steroids. It is also a site for their synthesis. Sex steroids in numerous ways can modify cerebral physiology, and along with many processes adult neurogenesis is also modulated by sex steroids. This review will focus on the effects of the main steroids, estrogens, androgens and progestogens, and unveil some aspects of their partly disclosed mechanisms of actions. Gonadal steroids act on different steps of neurogenesis: cell proliferation seems to be increased by estrogens only, while androgens and progestogens favor neuronal renewal by increasing cell survival; differentiation is a common target. Aging is characterized by a cognitive deficiency, paralleled by a decrease in the rate of neuronal renewal and in the levels of circulating gonadal hormones. Therefore, the effects of gonadal hormones on the aging brain are important to consider. The review will also be expanded to related molecules which are agonists to the nuclear receptors. Sex steroids can modify adult neuronal renewal and the extensive knowledge of their actions on neurogenesis is essential, as it can be a leading pathway to therapeutic perspectives.
Collapse
Affiliation(s)
- Christine Heberden
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.
| |
Collapse
|
37
|
Albin-Brooks C, Nealer C, Sabihi S, Haim A, Leuner B. The influence of offspring, parity, and oxytocin on cognitive flexibility during the postpartum period. Horm Behav 2017; 89:130-136. [PMID: 28062230 PMCID: PMC5986067 DOI: 10.1016/j.yhbeh.2016.12.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/23/2016] [Accepted: 12/31/2016] [Indexed: 11/25/2022]
Abstract
Pregnancy and the postpartum period are times of profound behavioral change including alterations in cognitive function. This has been most often studied using hippocampal-dependent tasks assessing spatial learning and memory. However, less is known about the cognitive effects of motherhood for tasks that rely on areas other than the hippocampus. We have previously shown that postpartum females perform better on the extradimensional phase of an attentional set shifting task, a measure of cognitive flexibility which is dependent on the medial prefrontal cortex (mPFC). The present experiments aimed to extend this work by examining the importance of postpartum stage as well as offspring and parity in driving improved mPFC cognitive function during motherhood. We also examined whether the neuropeptide oxytocin, which plays a role in regulating numerous maternal functions, mediates enhanced cognitive flexibility during motherhood. Our results demonstrate that compared to virgin females, cognitive flexibility is enhanced in mothers regardless of postpartum stage and is not affected by parity since both first (primiparous) and second (biparous) time mothers showed the enhancement. Moreover, we found that improved cognitive flexibility in mothers requires the presence of offspring, as removal of the pups abolished the cognitive enhancement in postpartum females. Lastly, using an oxytocin receptor antagonist, we demonstrate that oxytocin signaling in the mPFC is necessary for the beneficial effects of motherhood on cognitive flexibility. Together, these data provide insights into the temporal, experiential and hormonal factors which regulate mPFC-dependent cognitive function during the postpartum period.
Collapse
Affiliation(s)
| | - Connor Nealer
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Sara Sabihi
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Achikam Haim
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Benedetta Leuner
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Behavioral Neuroendocrinology Group, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
38
|
|
39
|
Colangelo LA, Ouyang P, Golden SH, Szklo M, Gapstur SM, Vaidya D, Liu K. Do sex hormones or hormone therapy modify the relation of n-3 fatty acids with incident depressive symptoms in postmenopausal women? The MESA Study. Psychoneuroendocrinology 2017; 75:26-35. [PMID: 27768981 PMCID: PMC5135581 DOI: 10.1016/j.psyneuen.2016.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/04/2016] [Accepted: 10/11/2016] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Considering that estradiol (E2) and n-3 polyunsaturated fatty acids (PUFAs) have roles in neurogenesis and in neurotransmission, we examined whether the association of PUFAs with incident depressive symptoms in postmenopausal women is modified by hormone therapy (HT) use or estrogen status. METHODS Women (N=1616) free of depressive symptoms at baseline (2000-2002) in the Multi-Ethnic Study of Atherosclerosis were classified by HT usage and quartiles of dietary eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and the sum EPA+DHA. Women with serum E2 ≤0.073 nmol/L (sample median), were classified low on E2. Poisson regression was used to model incident depressive symptoms at examination 3 (2004-05), defined by the Center for Epidemiological Studies Depression Scale ≥16 or taking an antidepressant, first as a function of HT use and n-3 PUFA quartiles, and second, as a function of low E2 status and n-3 PUFA quartiles. RESULTS Among HT non- users, positive, graded relationships (p-trends≤0.003) were found between PUFAs and incident depressive symptoms. Compared to the lowest quartile, the adjusted risk ratios (RRs) for the highest were 2.10, 2.39, and 2.04 for EPA, DHA, and EPA+DHA, respectively. For HT users, no associations were seen. When analyses were run for E2 status, the RRs over quartiles of the PUFAs were positive and graded for low E2 women, but were null for High E2 women. CONCLUSIONS Higher intakes of DHA and EPA were associated with higher risk of depressive symptoms in nonusers of HT, contrary to hypothesis.
Collapse
Affiliation(s)
- Laura A Colangelo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, 680 North Lake Shore Drive, Suite 1400, Chicago, IL 60611, United States.
| | - Pamela Ouyang
- Department of Medicine, Johns Hopkins University, Baltimore,
Maryland
| | | | - Moyses Szklo
- Department of Epidemiology, Johns Hopkins University, Baltimore,
Maryland
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta,
Georgia
| | - Dhananjay Vaidya
- Department of Medicine, Johns Hopkins University, Baltimore,
Maryland
| | - Kiang Liu
- Department of Preventive Medicine, Feinberg School of Medicine,
Northwestern University, 680 North Lake Shore Drive, Suite 1400, Chicago, Illinois
60611
| |
Collapse
|
40
|
Hoekzema E, Barba-Müller E, Pozzobon C, Picado M, Lucco F, García-García D, Soliva JC, Tobeña A, Desco M, Crone EA, Ballesteros A, Carmona S, Vilarroya O. Pregnancy leads to long-lasting changes in human brain structure. Nat Neurosci 2016; 20:287-296. [DOI: 10.1038/nn.4458] [Citation(s) in RCA: 331] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 11/15/2016] [Indexed: 12/14/2022]
|
41
|
How age, sex and genotype shape the stress response. Neurobiol Stress 2016; 6:44-56. [PMID: 28229108 PMCID: PMC5314441 DOI: 10.1016/j.ynstr.2016.11.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/19/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022] Open
Abstract
Exposure to chronic stress is a leading pre-disposing factor for several neuropsychiatric disorders as it often leads to maladaptive responses. The response to stressful events is heterogeneous, underpinning a wide spectrum of distinct changes amongst stress-exposed individuals'. Several factors can underlie a different perception to stressors and the setting of distinct coping strategies that will lead to individual differences on the susceptibility/resistance to stress. Beyond the factors related to the stressor itself, such as intensity, duration or predictability, there are factors intrinsic to the individuals that are relevant to shape the stress response, such as age, sex and genetics. In this review, we examine the contribution of such intrinsic factors to the modulation of the stress response based on experimental rodent models of response to stress and discuss to what extent that knowledge can be potentially translated to humans. Effect of age in the stress response. Effect of sex in the stress response. Effect of genotype in the stress response.
Collapse
|
42
|
Candeias E, Duarte AI, Sebastião I, Fernandes MA, Plácido AI, Carvalho C, Correia S, Santos RX, Seiça R, Santos MS, Oliveira CR, Moreira PI. Middle-Aged Diabetic Females and Males Present Distinct Susceptibility to Alzheimer Disease-like Pathology. Mol Neurobiol 2016; 54:6471-6489. [PMID: 27730513 DOI: 10.1007/s12035-016-0155-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/22/2016] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is a highly concerning public health problem of the twenty-first century. Currently, it is estimated that T2D affects 422 million people worldwide with a rapidly increasing prevalence. During the past two decades, T2D has been widely shown to have a major impact in the brain. This, together with the cognitive decline and increased risk for dementia upon T2D, may arise from the complex interaction between normal brain aging and central insulin signaling dysfunction. Among the several features shared between T2D and some neurodegenerative disorders (e.g., Alzheimer disease (AD)), the impairment of insulin signaling may be a key link. However, these may also involve changes in sex hormones' function and metabolism, ultimately contributing to the different susceptibilities between females and males to some pathologies. For example, female sex has been pointed as a risk factor for AD, particularly after menopause. However, less is known on the underlying molecular mechanisms or even if these changes start during middle-age (perimenopause). From the above, we hypothesized that sex differentially affects hormone-mediated intracellular signaling pathways in T2D brain, ultimately modulating the risk for neurodegenerative conditions. We aimed to evaluate sex-associated alterations in estrogen/insulin-like growth factor-1 (IGF-1)/insulin-related signaling, oxidative stress markers, and AD-like hallmarks in middle-aged control and T2D rat brain cortices. We used brain cortices homogenates obtained from middle-aged (8-month-old) control Wistar and non-obese, spontaneously T2D Goto-Kakizaki (GK) male and female rats. Peripheral characterization of the animal models was done by standard biochemical analyses of blood, plasma, or serum. Steroid sex hormones, oxidative stress markers, and AD-like hallmarks were given by specific ELISA kits and colorimetric techniques, whereas the levels of intracellular signaling proteins were determined by Western blotting. Albeit the high levels of plasma estradiol and progesterone observed in middle-aged control females suggested that they were still under their reproductive phase, some gonadal dysfunction might be already occurring in T2D ones, hence, anticipating their menopause. Moreover, the higher blood and lower brain cholesterol levels in female rats suggested that its dysfunctional uptake into the brain cortex may also hamper peripheral estrogen uptake and/or its local brain steroidogenic metabolism. Despite the massive drop in IGF-1 levels in females' brains, particularly upon T2D, they might have developed some compensatory mechanisms towards the maintenance of estrogen, IGF-1, and insulin receptors function and of the subsequent Akt- and ERK1/2-mediated signaling. These may ultimately delay the deleterious AD-like brain changes (including oxidative damage to lipids and DNA, amyloidogenic processing of amyloid precursor protein and increased tau protein phosphorylation) associated with T2D and/or age (reproductive senescence) in female rats. By demonstrating that differential sex steroid hormone profiles/action may play a pivotal role in brain over T2D progression, the present study reinforces the need to establish sex-specific preventive and/or therapeutic approaches and an appropriate time window for the efficient treatment against T2D and AD.
Collapse
Affiliation(s)
- E Candeias
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - A I Duarte
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal.
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal.
| | - I Sebastião
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
| | - M A Fernandes
- Life Sciences Department, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal
- Instituto do Mar, Life Sciences Department, University of Coimbra, 3004-517, Coimbra, Portugal
| | - A I Plácido
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - C Carvalho
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - S Correia
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - R X Santos
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Life Sciences Department, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal
| | - R Seiça
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - M S Santos
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Instituto do Mar, Life Sciences Department, University of Coimbra, 3004-517, Coimbra, Portugal
| | - C R Oliveira
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute of Biochemistry, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - P I Moreira
- CNC- Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal.
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal.
| |
Collapse
|
43
|
Luo Y, Xiao Q, Chao F, He Q, Lv F, Zhang L, Gao Y, Qiu X, Huang C, Li Y, Wang S, Jiang R, Gu H, Tang Y. 17β-estradiol replacement therapy protects myelin sheaths in the white matter of middle-aged female ovariectomized rats: a stereological study. Neurobiol Aging 2016; 47:139-148. [PMID: 27592282 DOI: 10.1016/j.neurobiolaging.2016.07.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/29/2016] [Accepted: 07/31/2016] [Indexed: 12/28/2022]
Abstract
Many studies have shown that estrogen replacement therapy (ERT) can improve cognitive function and affect the structure of the brain, including the white matter, in postmenopausal women. However, it is unclear whether ERT plays an important role in white matter remodeling in postmenopausal women. In the present study, middle-aged (9-12-month-old) female Sprague-Dawley rats were bilaterally ovariectomized (OVX) and randomly allocated to the vehicle treatment (OVX+Veh) group or the 17β-estradiol replacement (OVX+E) group. After 1 month of treatment, spatial learning and memory capacities were assessed using the Morris water maze task. Then, stereological methods were used to quantitatively evaluate white matter volume and myelinated fiber parameters of the white matter in the 2 groups of rats. The results revealed that the mean escape latency of the OVX+E rats in the Morris water maze task was significantly shorter than that of the OVX+Veh rats. The volume density of the myelinated fibers and the volume density and total volume of the myelin sheaths were significantly greater in the OVX+E rats than in the OVX+Veh rats. However, there were no significant differences in white matter volume or in the total length or volume of myelinated fibers in white matter between the 2 groups of rats. Our results showed that 1 month of ERT had significant beneficial effects on spatial learning capacity and on the myelin sheaths and myelinated fibers in the white matter of middle-aged OVX rats.
Collapse
Affiliation(s)
- Yanmin Luo
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Ministry of Education, Chongqing Medical University, Chongqing, PR China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Qian Xiao
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Ministry of Education, Chongqing Medical University, Chongqing, PR China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Fenglei Chao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Qi He
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Fulin Lv
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Lei Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Yuan Gao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Xuan Qiu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Chunxia Huang
- Department of Physiology, Chongqing Medical University, Chongqing, PR China
| | - Yongde Li
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Sanrong Wang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Rong Jiang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Hengwei Gu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
44
|
Hamson DK, Roes MM, Galea LAM. Sex Hormones and Cognition: Neuroendocrine Influences on Memory and Learning. Compr Physiol 2016; 6:1295-337. [DOI: 10.1002/cphy.c150031] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Mahmoud R, Wainwright SR, Galea LAM. Sex hormones and adult hippocampal neurogenesis: Regulation, implications, and potential mechanisms. Front Neuroendocrinol 2016; 41:129-52. [PMID: 26988999 DOI: 10.1016/j.yfrne.2016.03.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 11/16/2022]
Abstract
Neurogenesis within the adult hippocampus is modulated by endogenous and exogenous factors. Here, we review the role of sex hormones in the regulation of adult hippocampal neurogenesis in males and females. The review is framed around the potential functional implications of sex hormone regulation of adult hippocampal neurogenesis, with a focus on cognitive function and mood regulation, which may be related to sex differences in incidence and severity of dementia and depression. We present findings from preclinical studies of endogenous fluctuations in sex hormones relating to reproductive function and ageing, and from studies of exogenous hormone manipulations. In addition, we discuss the modulating roles of sex, age, and reproductive history on the relationship between sex hormones and neurogenesis. Because sex hormones have diverse targets in the central nervous system, we overview potential mechanisms through which sex hormones may influence hippocampal neurogenesis. Lastly, we advocate for a more systematic consideration of sex and sex hormones in studying the functional implications of adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Rand Mahmoud
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Steven R Wainwright
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Liisa A M Galea
- Department of Psychology, University of British Columbia, Vancouver, Canada; Centre for Brain Health, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
46
|
Korol DL, Pisani SL. Estrogens and cognition: Friends or foes?: An evaluation of the opposing effects of estrogens on learning and memory. Horm Behav 2015; 74:105-15. [PMID: 26149525 PMCID: PMC4573330 DOI: 10.1016/j.yhbeh.2015.06.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/19/2015] [Accepted: 06/25/2015] [Indexed: 12/19/2022]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Estrogens are becoming well known for their robust enhancement on cognition particularly for learning and memory that relies upon functioning of the hippocampus and related neural systems. What is also emerging is that estrogen modulation of cognition is not uniform, at times enhancing yet at other times impairing learning. This review explores the bidirectional effects of estrogens on learning from a multiple memory systems view, focusing on the hippocampus and striatum, whereby modulation by estrogens sorts according to task attributes and neural systems engaged during cognition. We highlight our findings showing that the ability to solve hippocampus-sensitive tasks typically improves under relatively high estrogen status while the ability to solve striatum-sensitive tasks degrades with estrogen exposures. Though constrained by dose and timing of exposure, these opposing enhancements and impairments of cognition can be observed following treatments with different estrogenic compounds including the hormone estradiol, the isoflavone genistein found in soybeans, and agonists that are selective for specific estrogen receptors, suggesting that activation of a single receptor type is sufficient to produce the observed shifts in learning strategies. Using this multi-dimensional framework will allow us to extend our thinking of the relationship between estrogens and cognition to other brain regions and cognitive functions.
Collapse
Affiliation(s)
- Donna L Korol
- Department of Biology, Syracuse University, Department of Neuroscience and Physiology, SUNY-Upstate Medical University, Syracuse, NY 13244, USA.
| | - Samantha L Pisani
- Neuroscience Program and Medical Scholars Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
47
|
Duarte-Guterman P, Yagi S, Chow C, Galea LAM. Hippocampal learning, memory, and neurogenesis: Effects of sex and estrogens across the lifespan in adults. Horm Behav 2015; 74:37-52. [PMID: 26122299 DOI: 10.1016/j.yhbeh.2015.05.024] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/29/2015] [Accepted: 05/26/2015] [Indexed: 01/12/2023]
Abstract
This article is part of a Special Issue "Estradiol and Cognition". There are sex differences in hippocampus-dependent cognition and neurogenesis suggesting that sex hormones are involved. Estrogens modulate certain forms of spatial and contextual memory and neurogenesis in the adult female rodent, and to a lesser extent male, hippocampus. This review focuses on the effects of sex and estrogens on hippocampal learning, memory, and neurogenesis in the young and aged adult rodent. We discuss how factors such as the type of estrogen, duration and dose of treatment, timing of treatment, and type of memory influence the effects of estrogens on cognition and neurogenesis. We also address how reproductive experience (pregnancy and mothering) and aging interact with estrogens to modulate hippocampal cognition and neurogenesis in females. Given the evidence that adult hippocampal neurogenesis plays a role in long-term spatial memory and pattern separation, we also discuss the functional implications of regulating neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Paula Duarte-Guterman
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Shunya Yagi
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Carmen Chow
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Liisa A M Galea
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada.
| |
Collapse
|