1
|
Lee BH, Eid RS, Hodges TE, Barth C, Galea LAM. Leveraging research into sex differences and steroid hormones to improve brain health. Nat Rev Endocrinol 2025; 21:214-229. [PMID: 39587332 DOI: 10.1038/s41574-024-01061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
Sex differences, driven in part by steroid hormones, shape the structure and function of the brain throughout the lifespan and manifest across brain health and disease. The influence of steroid hormones on neuroplasticity, particularly in the adult hippocampus, differs between the sexes, which has important implications for disorders and diseases that compromise hippocampus integrity, such as depression and Alzheimer disease. This Review outlines the intricate relationship between steroid hormones and hippocampal neuroplasticity across the adult lifespan and explores how the unique physiology of male and female individuals can affect health and disease. Despite calls to include sex and gender in research, only 5% of neuroscience studies published in 2019 directly investigated the influence of sex. Drawing on insights from depression, Alzheimer disease and relevant hippocampal plasticity, this Review underscores the importance of considering sex and steroid hormones to achieve a comprehensive understanding of disease susceptibility and mechanisms. Such consideration will enable the discovery of personalized treatments, ultimately leading to improved health outcomes for all.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Rand S Eid
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Travis E Hodges
- Department of Psychology and Education, Mount Holyoke College, South Hadley, MA, USA
| | - Claudia Barth
- Division for Mental Health and Substance Abuse, Diakonhjemmet Hospital, Oslo, Norway
| | - Liisa A M Galea
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Zhang N, Yu M, Zhao Q, Feng B, Deng Y, Bean JC, Liu Q, Eappen BP, He Y, Conde KM, Liu H, Yang Y, Tu L, Wang M, Li Y, Yin N, Liu H, Han J, Threat DA, Xu N, Smiley T, Xu P, Chen L, Zeng T, He Y, Wang C. Altered thermal preference by preoptic estrogen receptor alpha neurons in postpartum females. Mol Metab 2025; 93:102108. [PMID: 39909189 PMCID: PMC11849645 DOI: 10.1016/j.molmet.2025.102108] [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: 12/05/2024] [Revised: 01/14/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
OBJECTIVE This study aims to investigate how reproductive experience (RE) alters thermal preference and thermoregulation in female mice, with a focus on estrogen receptor alpha (ERα)-expressing neurons in the preoptic area (POA). METHODS Thermal preference and body temperature were measured in female mice with and without RE, and virgin female mice with selective deletion of ERα from the POA (ERαPOA-KO). The number and activity of ERα-expressing POA neurons (ERαPOA) were assessed using immunohistochemistry and in vitro electrophysiology in response to temperature changes and ERα agonist. RESULTS We showed that female mice prefer a cooler environment starting from late pregnancy and persisting long term postpartum. Female mice with RE (>4 weeks post-weaning) displayed lower body temperature and a lower thermal preferred temperature, and lost preference for warm environments (30 °C) but preserved avoidance of cold environments (15 °C). This was associated with a significant decrease in the number of ERαPOA neurons. Importantly, virgin female ERαPOA-KO mice displayed lower thermal preferred temperature and impaired warm preference, mimicking RE mice. We further found that distinct ERαPOA subpopulations can be regulated by temperature changes with or without presynaptic blockers, and by ERα agonist. More importantly, RE decreased the number of warm-activated ERαPOA neurons and reduced the excitatory effects of warmth and estrogen-ERα signaling, while cold-activated ERαPOA neurons were slightly enhanced in female mice with RE. CONCLUSION Our results support that the thermosensing ability and estrogenic effects in ERαPOA neurons are regulated by reproductive experience, altering thermal preference.
Collapse
Affiliation(s)
- Nan Zhang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA; Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China
| | - Meng Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Qianru Zhao
- Pennington Biomedical Research Center, Brain Glycemic and Metabolism Control Department, Louisiana State University, Baton Rouge, LA, 70808, USA; Department of Biological Chemistry, School of Pharmaceutical Sciences, South-central Minzu University, Wuhan, 430074, China
| | - Bing Feng
- Pennington Biomedical Research Center, Brain Glycemic and Metabolism Control Department, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - Yue Deng
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Jonathan C Bean
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Qingzhuo Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Benjamin P Eappen
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yang He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Kristine M Conde
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Hailan Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Longlong Tu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Mengjie Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yongxiang Li
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Na Yin
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Hesong Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Junying Han
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Darah Ave Threat
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Nathan Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Taylor Smiley
- Pennington Biomedical Research Center, Brain Glycemic and Metabolism Control Department, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - Pingwen Xu
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China.
| | - Yanlin He
- Pennington Biomedical Research Center, Brain Glycemic and Metabolism Control Department, Louisiana State University, Baton Rouge, LA, 70808, USA.
| | - Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
Pestana JE, Graham BM. The impact of estrous cycle on anxiety-like behaviour during unlearned fear tests in female rats and mice: A systematic review and meta-analysis. Neurosci Biobehav Rev 2024; 164:105789. [PMID: 39002829 DOI: 10.1016/j.neubiorev.2024.105789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024]
Abstract
Anxiety fluctuates across the human menstrual cycle, with symptoms worsening during phases of declining or low ovarian hormones. Similar findings have been observed across the rodent estrous cycle, however, the magnitude and robustness of these effects have not been meta-analytically quantified. We conducted a systematic review and meta-analysis of estrous cycle effects on anxiety-like behaviour (124 articles; k = 259 effect sizes). In both rats and mice, anxiety-like behaviour was higher during metestrus/diestrus (lower ovarian hormones) than proestrus (higher ovarian hormones) (g = 0.44 in rats, g = 0.43 in mice). There was large heterogeneity in the data, which was partially accounted for by strain, experimental task, and reproductive status. Nonetheless, the effect of estrous cycle on anxiety-like behaviour was highly robust, with the fail-safe N test revealing the effect would remain significant even if 21,388 additional studies yielded null results. These results suggest that estrous cycle should be accounted for in studies of anxiety in females. Doing so will facilitate knowledge about menstrual-cycle regulation of anxiety disorders in humans.
Collapse
|
4
|
Faraji M, Viera-Resto OA, Setlow B, Bizon JL. Effects of reproductive experience on cost-benefit decision making in female rats. Front Behav Neurosci 2024; 18:1304408. [PMID: 38352625 PMCID: PMC10863065 DOI: 10.3389/fnbeh.2024.1304408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024] Open
Abstract
Many individuals undergo mating and/or other aspects of reproductive experience at some point in their lives, and pregnancy and childbirth in particular are associated with alterations in the prevalence of several psychiatric disorders. Research in rodents shows that maternal experience affects spatial learning and other aspects of hippocampal function. In contrast, there has been little work in animal models concerning how reproductive experience affects cost-benefit decision making, despite the relevance of this aspect of cognition for psychiatric disorders. To begin to address this issue, reproductively experienced (RE) and reproductively naïve (RN) female Long-Evans rats were tested across multiple tasks that assess different forms of cost-benefit decision making. In a risky decision-making task, in which rats chose between a small, safe food reward and a large food reward accompanied by variable probabilities of punishment, RE females chose the large risky reward significantly more frequently than RN females (greater risk taking). In an intertemporal choice task, in which rats chose between a small, immediate food reward and a large food reward delivered after a variable delay period, RE females chose the large reward less frequently than RN females. Together, these results show distinct effects of reproductive experience on different forms of cost-benefit decision making in female rats, and highlight reproductive status as a variable that could influence aspects of cognition relevant for psychiatric disorders.
Collapse
Affiliation(s)
- Mojdeh Faraji
- Department of Psychiatry, University of Florida, Gainesville, FL, United States
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, United States
| | - Omar A. Viera-Resto
- Department of Psychiatry, University of Florida, Gainesville, FL, United States
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL, United States
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, United States
- McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Jennifer L. Bizon
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, United States
- McKnight Brain Institute, University of Florida, Gainesville, FL, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Pestana JE, Graham BM. Reproductive experience alters the effects of diazepam and fluoxetine on anxiety-like behaviour, fear extinction, and corticosterone levels in female rats. Psychopharmacology (Berl) 2023; 240:2515-2528. [PMID: 37581635 PMCID: PMC10640474 DOI: 10.1007/s00213-023-06446-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
OVERVIEW Reproductive experience (pregnancy and motherhood) leads to long-term changes in the neurobiological and hormonal features of anxiety in rats and humans. The aim of this study was to examine whether reproductive experience alters the effects of two pharmacological treatments for anxiety, a benzodiazepine (diazepam) and a selective serotonin reuptake inhibitor (fluoxetine), on animal models of anxiety. METHODS In Experiment 1, virgin (n = 47) and age-matched mother (n = 50) rats at 1-month post-weaning were injected with diazepam (1.3 mg/kg or 1.7 mg/kg, i.p.) or vehicle, in the proestrus (high estradiol/progesterone/allopregnanolone) or metestrus (low estradiol/progesterone/allopregnanolone) phase of the estrous cycle 30 min prior to the elevated plus maze (EPM). In Experiment 2, virgin (n = 25) and mother rats (n = 20) were administered fluoxetine (10 mg/kg) or vehicle for 2 weeks prior to being tested on a Pavlovian fear conditioning and extinction protocol, and the EPM. RESULTS Replicating past research, in virgin rats, the low dose of diazepam produced anxiolytic-like effects in proestrus, but only the high dose was anxiolytic-like in metestrus. In contrast, in mother rats, both doses of diazepam were anxiolytic-like irrespective of estrous phase. Fluoxetine produced anxiogenic-like effects in virgin rats during fear extinction and the EPM, but had no behavioural effects in mothers. In contrast, fluoxetine increased plasma corticosterone levels measured 30-min post-EPM in mothers, but not virgin rats. CONCLUSIONS Reproductive experience alters the dose responsivity and efficacy of common anti-anxiety medications in female rats. These findings highlight the importance of considering reproductive status in studies on anxiety and its treatment.
Collapse
Affiliation(s)
- Jodie E Pestana
- School of Psychology, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Bronwyn M Graham
- School of Psychology, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
6
|
Pestana JE, Kershaw KA, Graham BM. The impact of the ovarian cycle on anxiety, allopregnanolone, and corticotropin releasing hormone changes after motherhood in female rats and women. Transl Psychiatry 2023; 13:183. [PMID: 37253788 DOI: 10.1038/s41398-023-02480-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/02/2023] [Accepted: 05/15/2023] [Indexed: 06/01/2023] Open
Abstract
Fluctuations in ovarian steroids across the estrous and menstrual cycle in female rats and women, respectively, are associated with changes in anxiety. Pregnancy causes long-term changes to ovarian hormone release, yet research on estrous- and menstrual-related changes in anxiety has focused on reproductively inexperienced females. Therefore, this study assessed whether the impact of estrous and menstrual cycles on anxiety differs pre- versus post-motherhood in female rats (n = 32) and a community sample of women (n = 63). Estrous cycle phase altered anxiety-like behavior in virgin rats, but had no effect in age-matched mother rats tested 1-month post-weaning. In humans, menstrual cycle phase was associated with ecological momentary assessed anxiety and mood in non-mothers, but not mothers; although, the menstrual cycle × reproductive status interaction for anxiety, but not mood, was rendered non-significant with age and cycle length as covariates. These findings suggest that changes in anxiety coincident with cycling hormones is an evolutionarily conserved feature of the estrous and menstrual cycle in rats and women, which is mitigated following motherhood in both species. We identified several potential mechanisms for the observed dissociation in estrous cycle effects on anxiety. Compared to virgin rats, mother rats had a lower peak and blunted decline in circulating allopregnanolone during proestrus, upregulated GABAA receptor subunit (α1, α2, α5, α4, ß2) mRNA in the ventral hippocampus, and altered corticotropin-releasing hormone mRNA across the estrous cycle in the basolateral amygdala. Together, these findings suggest that the mechanisms underlying anxiety regulation undergo fundamental transformation following pregnancy in female rats and humans.
Collapse
Affiliation(s)
- Jodie E Pestana
- School of Psychology, UNSW Sydney, Sydney, NSW, 2052, Australia.
| | - Kelly A Kershaw
- School of Psychology, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | |
Collapse
|
7
|
Hernández-Hernández OT, Herrera-Pérez JJ, Chavira R, Lemini C, Martínez-Mota L. Effects of post-ovariectomy time frame and age on the antidepressant-like actions of estradiol and prolame in female rats. Horm Behav 2022; 141:105154. [PMID: 35306314 DOI: 10.1016/j.yhbeh.2022.105154] [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: 07/12/2021] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 11/23/2022]
Abstract
Estrogen replacement therapy (ERT) is an effective treatment for symptoms associated with climacteric and depression some women experience during perimenopause and menopause. The antidepressant-like effects of ERT may depend on the type of estrogen, age, and time when restitution is initiated after hormonal decline. Prolame is a synthetic steroid with estrogenic and antidepressant-like effects that may produce fewer adverse effects. We hypothesize that such actions of prolame on females depend on age and the duration of hormone deprivation period. We assessed the antidepressant-like effects of 17β-estradiol (E2) and prolame in young and middle-aged rats across different post-ovariectomy (Ovx) time frames. Independent groups of young adults and middle-aged female rats were tested in the forced swimming test (FST) at 3, 8, 16, and 24 weeks post-Ovx. Prolame and E2 were administered in a sub-chronic schedule consisting of three injections before the FST. Likewise, the utero-trophic effects of these hormones were analyzed. We found that E2 and prolame reduced immobility in young rats 3 and 8 weeks after Ovx; in contrast, only prolame produced this effect in middle-aged rats three weeks post-Ovx. E2 and prolame increased the animals' utero-somatic index at all post-Ovx times, but the action of E2 and prolame produced a greater response in young adult rats. Our findings showed that the antidepressant-like effects of E2 and prolame depend on the post-Ovx time frame, age, and estrogen type. Interestingly, our results indicate that, in contrast to E2, prolame maintained its antidepressant effect in middle-aged rats.
Collapse
Affiliation(s)
- Olivia Tania Hernández-Hernández
- Consejo Nacional de Ciencia y Tecnología Research Fellow, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Delegación Tlalpan, 14370 Ciudad de México, Mexico
| | - José Jaime Herrera-Pérez
- Laboratorio de Farmacología Conductual, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Delegación Tlalpan, 14370 Ciudad de México, Mexico
| | - Roberto Chavira
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, 14080 Ciudad de México, Mexico
| | - Cristina Lemini
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad No. 3000, Ciudad Universitaria, Delegación Coyoacán, 04510 Ciudad de México, Mexico.
| | - Lucía Martínez-Mota
- Laboratorio de Farmacología Conductual, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Delegación Tlalpan, 14370 Ciudad de México, Mexico.
| |
Collapse
|
8
|
Farrar VS, Ramirez AV, Calisi RM. Effects of Parental Experience and Age On Expression of Prolactin, Vasoactive Intestinal Peptide and Their Receptors in a Biparental Bird (Columba Livia). Integr Comp Biol 2022; 62:30-40. [PMID: 35438167 DOI: 10.1093/icb/icac017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
As animals gain parental experience, they often show more rapid and efficient parental care responses that likely improve offspring survival and fitness. Changes in circulating hormones that underlie reproductive behaviors, including prolactin, have been found to correlate with parental experience in birds and mammals. Altered responsiveness to prolactin in key behavioral centers of the brain may also underlie the effects of experience on parental behaviors. Further, experience may also affect responsiveness to prolactin stimulatory hormones, such as hypothalamic vasoactive intestinal peptide (VIP). While experience has been shown to upregulate neural prolactin receptors and responsiveness in rodents, its effects on prolactin receptor gene expression remain unstudied in birds. To address this, we examined gene expression of pituitary prolactin, hypothalamic prolactin receptors in the preoptic area, hypothalamic VIP, and pituitary VIP receptors in both sexes of the biparental rock dove (Columba livia) when birds were not actively nesting. As age and parental experience are often confounded (i.e.,experienced parents tend to be older than their inexperienced counterparts), we measured gene expression in birds of varying combinations of age (0.6-3 years) and prior reproductive experience (0-12 chicks raised). We found that increasing experience with chicks correlated with lower PRLR expression in the preoptic area, and age correlated with lower VIP expression in birds of both sexes. Pituitary PRL and VIPR expression was not associated with parental experience or age. These results suggest there may be persistent effects of experience and age on neural responsiveness to, and regulation of, prolactin in birds.
Collapse
Affiliation(s)
- Victoria S Farrar
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616
| | - Alison V Ramirez
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616
| | - Rebecca M Calisi
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616
| |
Collapse
|
9
|
Pestana JE, McCutcheon TB, Harmon-Jones SK, Richardson R, Graham BM. Maternal Experience Does Not Predict Fear Extinction and Anxiety-Like Behaviour in Primiparous Rats Post-weaning. Front Glob Womens Health 2022; 2:742337. [PMID: 34977862 PMCID: PMC8718406 DOI: 10.3389/fgwh.2021.742337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
Reproductive experience leads to long-lasting changes in anxiety-like behaviour and fear extinction, the laboratory model of exposure therapy for anxiety disorders. For example, fear extinction is influenced by estrous cycle in nulliparous (no reproductive experience) female rats, but this effect is abolished in primiparous (one reproductive experience) females. It is unclear whether such changes are driven by pregnancy, maternal experience of caring for offspring during the postpartum period, or a combination of both experiences. The present study sought to determine the influence of maternal experience (i.e., exposure to pups and mother-pup interactions) on fear extinction in primiparous rats. In Experiment 1, we tested whether pup exposure is necessary to mitigate estrous effects on fear extinction in primiparous rats. Age-matched nulliparous rats, primiparous rats, and primiparous rats who experienced pregnancy but not pup exposure, underwent fear conditioning on day 1 (2 months post-parturition), extinction training during proestrus (high sex hormones) or metestrus (low sex hormones) on day 2, and extinction recall on day 3. Replicating past research, nulliparous rats showed impaired extinction recall when they were extinguished during metestrus compared to proestrus. In contrast, primiparous rats with and without pup exposure showed comparable extinction recall irrespective of estrous phase. In Experiment 2, we assessed whether naturally-occurring variation in mother-pup interactions predict future fear extinction performance and anxiety-like behaviour. During the first week of lactation, primiparous rats were measured for maternal behaviours toward pups. Primiparous rats were then tested on the light-dark box and elevated plus maze to measure anxiety-like behaviour and underwent a fear extinction protocol 1 month post-weaning. We found no significant correlations between maternal behaviour and fear extinction outcomes or anxiety-like behaviour. Our findings suggest that pregnancy, not maternal experience, mitigates the impact of estrous cycle on fear extinction. In addition, natural variation in maternal experience does not appear to contribute to variability in future fear extinction outcomes or anxiety-like behaviour in primiparous rats.
Collapse
Affiliation(s)
- Jodie E Pestana
- School of Psychology, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Tayla B McCutcheon
- School of Psychology, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Sylvia K Harmon-Jones
- School of Psychology, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Rick Richardson
- School of Psychology, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| | - Bronwyn M Graham
- School of Psychology, University of New South Wales (UNSW) Sydney, Sydney, NSW, Australia
| |
Collapse
|
10
|
Effect of eCG in a short-term synchronization treatment on ovarian status, estrus synchrony, and ovulation in dairy goats managed under tropical conditions. Trop Anim Health Prod 2021; 53:246. [PMID: 33821404 DOI: 10.1007/s11250-021-02699-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/29/2021] [Indexed: 10/21/2022]
Abstract
The aim of this study was to assess the need of using eCG on short-term estrus synchronization protocol in nulliparous (NUL) and multiparous (MULT) dairy goats during the breeding season. Alpine (n = 20), Nubian (n = 20), and Saanen (n = 16) goats received 60 mg medroxyprogesterone acetate intravaginal sponges for 6 days plus 30 μg d-cloprostenol and 200 IU eCG (G-eCG, n = 28) or saline (G-Control, n = 28) 24 h before sponge removal. The NUL and MULT goats of each breed were equally assigned into the two treatments. Transrectal ultrasonography was used to evaluate ovulatory parameters, and teaser goats were used for estrus detection every 12 h from sponge removal to ovulation. eCG did not affect (P > 0.05) estrus response (~86%), diameter of ovulatory follicles (~6.8 mm), and number of ovulations (~1.6). Nevertheless, eCG led to earlier (P < 0.05) ovulation (G-eCG = 65.1 and G-Control = 73.2 h) and increased (P < 0.05) the ovulation rate (G-eCG = 96.4% and G-Control = 67.9%). In the absence of eCG, no differences regarding reproductive parameters (P > 0.05) were found between parity orders. Alpine MULT goats underwent a superior (P < 0.05) number of ovulations (2.2) in comparison to NUL goats (1.3). In conclusion, the exclusion of eCG from short-term estrus synchronization protocol did not interfere with estrus response but decreased the ovulation rate.
Collapse
|
11
|
Lawrence RH, Palumbo MC, Freeman SM, Guoynes CD, Bales KL. Developmental Fluoxetine Exposure Alters Behavior and Neuropeptide Receptors in the Prairie Vole. Front Behav Neurosci 2020; 14:584731. [PMID: 33304247 PMCID: PMC7701284 DOI: 10.3389/fnbeh.2020.584731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/23/2020] [Indexed: 12/28/2022] Open
Abstract
Developmental exposure to selective serotonin reuptake inhibitor (SSRI) increases the risk of Autism Spectrum Disorder (ASD), however, the underlying neurobiology of this effect is not fully understood. Here we used the socially monogamous prairie vole as a translational model of developmental SSRI exposure. Paired female prairie voles (n = 20) were treated with 5 mg/kg subcutaneous fluoxetine (FLX) or saline (SAL) daily from birth of the second litter until the day of birth of the 4th litter. This design created three cohorts of FLX exposure: postnatal exposure in litter 2, both prenatal and postnatal exposure in litter 3, and prenatal exposure in litter 4. Post-weaning, subjects underwent behavioral testing to detect changes in sociality, repetitive behavior, pair-bond formation, and anxiety-like behavior. Quantitative receptor autoradiography was performed for oxytocin, vasopressin 1a, and serotonin 1a receptor density in a subset of brains. We observed increased anxiety-like behavior and reduced sociality in developmentally FLX exposed adults. FLX exposure decreased oxytocin receptor binding in the nucleus accumbens core and central amygdala, and vasopressin 1a receptor binding in the medial amygdala. FLX exposure did not affect serotonin 1A receptor binding in any areas examined. Changes to oxytocin and vasopressin receptors may underlie the behavioral changes observed and have translational implications for the mechanism of the increased risk of ASD subsequent to prenatal SSRI exposure.
Collapse
Affiliation(s)
- Rebecca H Lawrence
- Department of Psychology, University of California, Davis, Davis, CA, United States.,California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Michelle C Palumbo
- California National Primate Research Center, University of California, Davis, Davis, CA, United States.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Sara M Freeman
- Department of Psychology, University of California, Davis, Davis, CA, United States.,California National Primate Research Center, University of California, Davis, Davis, CA, United States.,Department of Biology, Utah State University, Logan, UT, United States
| | - Caleigh D Guoynes
- Department of Psychology, University of California, Davis, Davis, CA, United States.,Department of Psychology, University of Wisconsin, Madison, WI, United States
| | - Karen L Bales
- Department of Psychology, University of California, Davis, Davis, CA, United States.,California National Primate Research Center, University of California, Davis, Davis, CA, United States.,Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, United States
| |
Collapse
|
12
|
Tang S, Graham BM. Hormonal, reproductive, and behavioural predictors of fear extinction recall in female rats. Horm Behav 2020; 121:104693. [PMID: 31981581 DOI: 10.1016/j.yhbeh.2020.104693] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/13/2020] [Accepted: 01/18/2020] [Indexed: 01/09/2023]
Abstract
The prevalence, severity and chronicity of anxiety disorders is significantly higher in women compared to men. Exposure therapy, the gold-standard treatment for anxiety disorders, can be modelled in the laboratory through Pavlovian fear extinction. Understanding the factors that influence fear extinction in females may aid in optimising the treatment of anxiety disorders in this population. The aim of the current study was therefore to explore the hormonal, reproductive and behavioural predictors of fear extinction recall in female rats by analysing data from nine published experiments that examined fear extinction in female rats. A hierarchical multiple regression analysis revealed that estrous cycle effects on extinction recall may be modulated by reproductive status. While the estrous phase in which nulliparous (virgin) rats undergo extinction training was predictive of extinction recall, no relationship between estrous phase and extinction recall was found among primiparous (one prior reproductive experience) rats. Moreover, estrous cycle predicted the relationship between early extinction and extinction recall in nulliparous rats, but not primiparous rats. Although reproductive status did not predict extinction recall, primiparous rats exhibited poor extinction recall relative to nulliparous rats extinguished during proestrus, and better extinction recall than nulliparous rats extinguished during metestrus. A faster rate of extinction, and lower fear responses at the end of extinction training were predictive of lower levels of CS-elicited fear during extinction recall in both nulliparous and primiparous female rats, while the length of extinction training was not predictive of extinction recall. The potential theoretical and clinical implications of these findings are discussed.
Collapse
Affiliation(s)
- Samantha Tang
- School of Psychology, UNSW Sydney, NSW 2052, Australia.
| | | |
Collapse
|
13
|
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: 5.6] [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
|
14
|
The Role of Hormonal and Reproductive Status in the Treatment of Anxiety Disorders in Women. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32002944 DOI: 10.1007/978-981-32-9705-0_26] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Exposure therapy, a key treatment for anxiety disorders, can be modelled in the laboratory using Pavlovian fear extinction. Understanding the hormonal and neurobiological mechanisms underlying fear extinction in females, who are twice more likely than males to present with anxiety disorders, may aid in optimising exposure therapy outcomes in this population. This chapter will begin by discussing the role of the sex hormones, estradiol and progesterone, in fear extinction in females. We will also propose potential mechanisms by which these hormones may modulate fear extinction. The second half of this chapter will discuss the long-term hormonal, neurological and behavioural changes that arise from pregnancy and motherhood and how these changes may alter the features of fear extinction in females. Finally, we will discuss implications of this research for the treatment of anxiety disorders in women with and without prior reproductive experience.
Collapse
|
15
|
Tang S, Graham BM. d-Cycloserine and estradiol enhance fear extinction in nulliparous but not primiparous female rats. Neurobiol Learn Mem 2019; 166:107088. [PMID: 31513850 DOI: 10.1016/j.nlm.2019.107088] [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] [Received: 06/20/2019] [Revised: 08/14/2019] [Accepted: 09/07/2019] [Indexed: 01/16/2023]
Abstract
Female reproductive experience has been shown to alter the hormonal, neurobiological and behavioural features of fear extinction, which is the laboratory basis of exposure therapy. This raises uncertainties as to whether pharmacological agents that enhance fear extinction in reproductively inexperienced females are equally effective in reproductively experienced females. The aim of the current study was therefore to compare the effects of two pharmacological enhancers of fear extinction, d-cycloserine (DCS) and estradiol, between nulliparous (virgin) and primiparous (reproductively experienced) female rats. In Experiment 1, nulliparous and primiparous females received systemic administration of either DCS or saline immediately after extinction training, and were tested for extinction recall the following day. DCS enhanced extinction recall in nulliparous females that showed low levels of freezing at the end of extinction training, but not among those that showed high levels of freezing at the end of extinction training. DCS did not enhance fear extinction in primiparous females, regardless of their level of freezing at the end of extinction training. In Experiment 2, nulliparous and primiparous female rats received systemic administration of either estradiol or vehicle prior to extinction training. Estradiol enhanced extinction recall among nulliparous females, but not primiparous females. Increasing the dose of estradiol administered prior to extinction training did not alter the outcomes in primiparous females (Experiment 3). Together, these findings suggest that reproductive status may be an important individual difference factor associated with the response to pharmacological modulators of extinction in rats. The implications of these findings for the pharmacological augmentation of exposure therapy in clinical populations are discussed.
Collapse
Affiliation(s)
- Samantha Tang
- School of Psychology, UNSW Sydney, NSW 2052, Australia.
| | | |
Collapse
|
16
|
Tang S, Graham BM. Reproductive experience alters the involvement of N-methyl-D-aspartate receptors in fear extinction, but not fear conditioning, in female Sprague Dawley rats. Psychopharmacology (Berl) 2019; 236:251-264. [PMID: 29959460 DOI: 10.1007/s00213-018-4956-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/19/2018] [Indexed: 01/05/2023]
Abstract
Recently, evidence has emerged showing that the behavioural and hormonal features of fear extinction are altered as a result of reproductive experience in both rats and humans. The current set of experiments sought to determine whether reproductive experience also alters the molecular features of fear extinction. In adult male rats, it has been widely demonstrated that the activation of N-methyl-D-aspartate receptors (NMDAR) is essential for fear extinction. We therefore compared the involvement of NMDAR in fear extinction between nulliparous (virgin) and primiparous (reproductively experienced) female rats. Nulliparous and primiparous females received systemic administrations of either MK-801 (a non-competitive NMDAR antagonist) or saline prior to extinction training. MK-801 was found to impair extinction recall in nulliparous females, but not primiparous females. When the same dose of MK-801 was administered prior to conditioning, both groups of rats showed impaired recall of conditioning the following day. The results of these experiments indicate that the extinction, but not the acquisition of fear, may become NMDAR-independent following reproductive experience.
Collapse
Affiliation(s)
- Samantha Tang
- School of Psychology, UNSW, Sydney, NSW, 2052, Australia.
| | | |
Collapse
|
17
|
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: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
18
|
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: 2.9] [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]
|
19
|
Stanojević S, Ćuruvija I, Blagojević V, Petrović R, Prijić I, Vujić V. The involvement of estrogen receptors α and β in the in vitro effects of 17β-estradiol on secretory profile of peritoneal macrophages from naturally menopausal female and middle-aged male rats. Exp Gerontol 2018; 113:86-94. [PMID: 30287187 DOI: 10.1016/j.exger.2018.09.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 09/13/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022]
Abstract
The systemic and extra- gonadal levels of 17β-estradiol (E2) change during aging, and affect the expression of estrogen receptors (ERs) in the immune cells of both females and males. The age-related cessation of ovarian function in females, as well as the tissue-specific expression of enzyme aromatase (estrogen synthase which significantly rises with the advancing age) in both males and females, both determine the concentration of E2 to which immune cells may be exposed. The present study was set up to investigate the direct influence of E2 in vitro on the secretory profile of peritoneal macrophages from young and naturally menopausal female rats, and from young and middle-aged male rats. The involvement of receptor(s) responsible for mediating the effects of E2 in vitro was examined by use of antagonists specific for ERα or ERβ. Whereas in macrophages from young female rats E2 treatment diminished interleukin (IL)-1β secretion, it increased it in young males, and the middle-aged females. The in vitro E2 treatment increased tumor necrosis factor (TNF)-α release by macrophages from young rats of both sexes, while it increased macrophage IL-6 release independently of both sex and age. At the same time, E2 decreased hydrogen peroxide (H2O2) production in macrophages from females, and increased it in male rats of both ages, whereas it diminished nitric oxide (NO) release in all experimental groups. Inspite of the sex- and age-specific effects of E2 on macrophage urea release, E2 did not affect the NO/urea ratio in macrophages from female rats, and diminished it in macrophages from both young and middle-aged male rats. Independently of the sex and age, E2 stimulated the release of inflammatory cytokines predominantly via macrophage ERα, and inhibited the IL-1β release in young females via ERβ. In contrast, E2 increased macrophage H2O2 and urea production by activating ERβ, but diminished their release via ERα. Our study may contribute to better understanding of the complex role(s) that E2 may play in innate immunity during aging, and that are dependent of sex.
Collapse
Affiliation(s)
- Stanislava Stanojević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia.
| | - Ivana Ćuruvija
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Veljko Blagojević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Raisa Petrović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Ivana Prijić
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Vesna Vujić
- Department of Chemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
20
|
Acevedo N, Rubin BS, Schaeberle CM, Soto AM. Perinatal BPA exposure and reproductive axis function in CD-1 mice. Reprod Toxicol 2018; 79:39-46. [PMID: 29752986 DOI: 10.1016/j.reprotox.2018.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 11/30/2022]
Abstract
Perinatal Bisphenol-A (BPA) exposure reduces fertility and fecundity in mice. This study examined effects of early BPA exposure on activation of gonadotropin releasing hormone (GnRH) neurons in conjunction with a steroid-induced luteinizing hormone (LH) surge, characterized patterns of estrous cyclicity and fertility over time, and assessed the ovarian follicular reserve to further explore factors responsible for the reduced fertility we previously described in this model. The percent activated GnRH neurons was reduced in BPA-exposed females at 3-6 months, and periods of persistent proestrus were increased. These data suggest that perinatal exposure to BPA reduces GnRH neuronal activation required for the generation of the LH surge and estrous cyclicity. Assessments of anti-Müllerian hormone (AMH) levels failed to suggest a decline in the follicular reserve at the BPA exposure levels examined.
Collapse
Affiliation(s)
- Nicole Acevedo
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, 02111, USA, USA
| | - Beverly S Rubin
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, 02111, USA, USA
| | - Cheryl M Schaeberle
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, 02111, USA, USA
| | - Ana M Soto
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, 02111, USA, USA.
| |
Collapse
|
21
|
Milligan-Saville JS, Graham BM. Mothers do it differently: reproductive experience alters fear extinction in female rats and women. Transl Psychiatry 2016; 6:e928. [PMID: 27779622 PMCID: PMC5290346 DOI: 10.1038/tp.2016.193] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/12/2016] [Accepted: 08/17/2016] [Indexed: 01/06/2023] Open
Abstract
Fear extinction is the laboratory basis of exposure therapy for anxiety disorders. Recent findings have revealed that estradiol is necessary to the consolidation of extinction memories in females. These findings are based on studies conducted using virgin rats and young women whose reproductive history is unknown. We hypothesized that motherhood, which results in extensive endocrinological, neurobiological and behavioral changes, may lead to alterations in fear extinction in females. We used a cross-species translational approach to investigate the impact of reproductive experience on fear extinction and fear relapse in female rats (n=116) and women (n=64). Although freezing during extinction recall was associated with estrous cycle phase during extinction training in virgin rats, this association was mitigated in age-matched reproductively experienced rats, even when fear extinction occurred 3 months after pups had been weaned, and even though reproductively experienced rats exhibited attenuated serum estradiol levels. In addition, although serum estradiol levels predicted extinction recall in human women with no prior reproductive experience, no such association was found in women with children. Finally, although virgin rats displayed both renewal and reinstatement after fear extinction, these common relapse phenomena were absent in rats with reproductive experience. Together, these findings suggest that reproductive experience alters the endocrine and behavioral features of fear extinction in females long after the hormonal surges of pregnancy and lactation have diminished. These results highlight the need to incorporate both hormonal and reproductive status as important factors in current models of fear extinction in females.
Collapse
Affiliation(s)
- J S Milligan-Saville
- School of Psychology, The University of New South Wales Australia, Sydney, NSW, Australia
| | - B M Graham
- School of Psychology, The University of New South Wales Australia, Sydney, NSW, Australia,School of Psychology, The University of New South Wales Australia, Sydney, NSW 2052, Australia.
| |
Collapse
|
22
|
Glynn LM, Davis EP, Sandman CA, Goldberg WA. Gestational hormone profiles predict human maternal behavior at 1-year postpartum. Horm Behav 2016; 85:19-25. [PMID: 27427279 PMCID: PMC5929113 DOI: 10.1016/j.yhbeh.2016.07.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/17/2016] [Accepted: 07/13/2016] [Indexed: 12/26/2022]
Abstract
In many non-human species, including primates, gestational reproductive hormones play an essential role in the onset of maternal motivation and behaviors. We investigated the associations between prepartum estradiol and progesterone and maternal behavior at 1-year postpartum in 177 women. Blood was obtained at five gestational time points and an index of quality of maternal care was determined using a well-validated mother-child interaction protocol. Women who exhibited higher quality maternal care at 1-year postpartum were characterized by unique gestational profiles of estradiol, progesterone and the estrogen to progesterone ratio; specifically by slower accelerations and levels of these hormone trajectories beginning in midgestation. Further, it appeared that both fetal sex and parity moderated these findings, with first time mothers and mothers of females showing stronger associations. In sum, these data document persisting associations between prepartum hormone profiles and human maternal behavior. More broadly, these findings add to the growing literature highlighting the perinatal period as one of critical neurodevelopment in the lifespan of the human female.
Collapse
Affiliation(s)
- Laura M Glynn
- Department of Psychology, Chapman University, One University Dr., Orange, CA 92868, United States; Department of Psychiatry & Human Behavior, University of California, Irvine, 101 The City Dr., Building 3, Route 88, Orange, CA 92868, United States.
| | - Elysia Poggi Davis
- Department of Psychiatry & Human Behavior, University of California, Irvine, 101 The City Dr., Building 3, Route 88, Orange, CA 92868, United States; Department of Psychology, University of Denver, 2155 South Race St., Denver, CO 80210, United States
| | - Curt A Sandman
- Department of Psychiatry & Human Behavior, University of California, Irvine, 101 The City Dr., Building 3, Route 88, Orange, CA 92868, United States
| | - Wendy A Goldberg
- Department of Psychology and Social Behavior, University of California, Irvine, 5300 Social and Behavioral Sciences Gateway, Irvine, CA 92697, United States
| |
Collapse
|
23
|
Bridges RS. Long-term alterations in neural and endocrine processes induced by motherhood in mammals. Horm Behav 2016; 77:193-203. [PMID: 26388065 PMCID: PMC4724454 DOI: 10.1016/j.yhbeh.2015.09.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/09/2015] [Accepted: 09/11/2015] [Indexed: 01/23/2023]
Abstract
This article is part of a Special Issue "Parental Care". The reproductive experience of pregnancy, lactation and motherhood can significantly remodel the female's biological state, affecting endocrine, neuroendocrine, neural, and immunological processes. The brain, pituitary gland, liver, thymus, and mammary tissue are among the structures that are modified by reproductive experience. The present review that focuses on rodent research, but also includes pertinent studies in sheep and other species, identifies specific changes in these processes brought about by the biological states of pregnancy, parturition, and lactation and how the components of reproductive experience contribute to the remodeling of the maternal brain and organ systems. Findings indicate that prior parity alters key circulating hormone levels and neural receptor gene expression. Moreover, reproductive experience results in modifications in neural processes and glial support. The possible role of pregnancy-induced neurogenesis is considered in the context of neuroplasticity and behavior, and the effects of reproductive experience on maternal memory, i.e. the retention of maternal behavior, together with anxiety and learning are presented. Together, these sets of findings support the concept that the neural and biological state of the adult female is significantly and dramatically altered on a long-term basis by the experiences of parity and motherhood. Remodeling of the maternal brain and other biological systems is posited to help facilitate adaptations to environmental/ecological challenges as the female raises young and ages.
Collapse
Affiliation(s)
- Robert S Bridges
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, USA.
| |
Collapse
|
24
|
Liu JL, Zuo RJ, Peng Y, Fu YS. The Impact of Multiparity on Uterine Gene Expression and Decidualization in Mice. Reprod Sci 2015; 23:687-94. [PMID: 26494701 DOI: 10.1177/1933719115612131] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
It has been well established that a previous pregnancy exhibits a beneficial effect on the subsequent pregnancy. However, the underlying mechanisms have not been defined. We hypothesized that multiparity may affect decidualization process during early pregnancy. To test this hypothesis, we analyzed global gene changes associated with multiparity in the mouse uterus using RNA-sequencing (RNA-seq). We identified a total of 131 differentially expressed genes (fold change > 2 and false discovery rate < 0.05), of which 58 were downregulated and 73 genes were upregulated in the second pregnancy (SP) compared to the first pregnancy. Functional clustering analysis showed that genes involved in stress response were significantly enriched. Most importantly, a significant portion of differentially expressed genes, 14 genes or 10.7%, overlapped with the gene list associated with decidualization. Quantitative reverse transcription (RT) polymerase chain reaction (qRT-PCR) analysis confirmed a decreased expression of 4 genes (Klk1, kallikrein 1; H2-Eb1, histocompatibility 2 class II antigen E beta; Mmp7, matrix metallopeptidase 7; Pdpn, podoplanin) and an increase in expression of 2 genes (Thy1, thymus cell antigen 1; Ptgs2, prostaglandin-endoperoxide synthase 2) in SP. Beyond protein-coding genes, we also identified a differentially expressed long noncoding RNA AI506816. Our data provide new insights into the molecular mechanisms underlying the beneficial effect of multiparity.
Collapse
Affiliation(s)
- Ji-Long Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ru-Juan Zuo
- College of Life Science, Xiamen University, Xiamen, China
| | - Ying Peng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yong-Sheng Fu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
25
|
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: 138] [Impact Index Per Article: 13.8] [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
|
26
|
Breast Cancer Prevention: Lessons to be Learned from Mechanisms of Early Pregnancy–Mediated Breast Cancer Protection. Cancer Res 2015; 75:803-7. [DOI: 10.1158/0008-5472.can-14-2717] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Tikk K, Sookthai D, Johnson T, Dossus L, Clavel-Chapelon F, Tjønneland A, Olsen A, Overvad K, Baglietto L, Rinaldi S, Romieu I, Boeing H, Trichopoulou A, Lagiou P, Trichopoulos D, Masala G, Agnoli C, Tumino R, Sacerdote C, Mattiello A, Buckland G, Sánchez S, Molina-Montes E, Amiano P, Castaño JMH, Barricarte A, Bueno-de-Mesquita HB, Monninkhof EM, Onland-Moret NC, Idahl A, Lundin E, Weiderpass E, Lund E, Waaseth M, Khaw KT, Key TJ, Travis RC, Gunter MJ, Riboli E, Kaaks R. Prolactin determinants in healthy women: A large cross-sectional study within the EPIC cohort. Cancer Epidemiol Biomarkers Prev 2014; 23:2532-42. [PMID: 25143360 DOI: 10.1158/1055-9965.epi-14-0613] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Experimental and epidemiologic data suggest that higher circulating prolactin is associated with breast cancer risk; however, how various risk factors for breast cancer influence prolactin levels in healthy women is not clear. METHODS We analyzed cross-sectional associations between several suggested reproductive and lifestyle risk factors for breast cancer and circulating prolactin among pre- and postmenopausal women, taking into account the use of current postmenopausal hormone therapy, among 2,560 controls from a breast cancer nested case-control study within the EPIC cohort. RESULTS Adjusted geometric mean prolactin levels were significantly higher among premenopausal women, and among postmenopausal women using hormone therapy compared with nonusers (8.2, 7.0, and 6.3 ng/mL, respectively; Pcat = <0.0001). Furthermore, prolactin levels were significantly higher among users of combined estrogen-progestin hormone therapy compared with users of estrogen-alone hormone therapy (6.66 vs. 5.90 ng/mL; Pcat = 0.001). Prolactin levels were lower among parous women compared with nulliparous women (8.61 vs. 10.95 ng/mL; Pcat = 0.0002, premenopausal women); the magnitude of this difference depended on the number of full-term pregnancies (22.1% lower, ≥3 vs. 1 pregnancy, Ptrend = 0.01). Results for parity were similar but lower in magnitude among postmenopausal women. Prolactin did not vary by other studied factors, with the exception of lower levels among postmenopausal smokers compared with never smokers. CONCLUSIONS Our study shows that current hormone therapy use, especially the use of combined hormone therapy, is associated with higher circulating prolactin levels in postmenopausal women, and confirms prior findings of lower circulating prolactin in parous women. IMPACT Our study extends the knowledge linking various breast cancer risk factors with circulating prolactin.
Collapse
Affiliation(s)
- Kaja Tikk
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Disorn Sookthai
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Theron Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laure Dossus
- Nutrition, Hormones, and Women's Health Team, Centre for Research in Epidemiology and Population Health (CESP), INSERM U1018, Villejuif, France. Univ Paris Sud, UMRS 1018, Villejuif, France. IGR, Villejuif, France
| | - Françoise Clavel-Chapelon
- Nutrition, Hormones, and Women's Health Team, Centre for Research in Epidemiology and Population Health (CESP), INSERM U1018, Villejuif, France. Univ Paris Sud, UMRS 1018, Villejuif, France. IGR, Villejuif, France
| | | | - Anja Olsen
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Kim Overvad
- Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Laura Baglietto
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia. Centre for Molecular, Environmental, Genetic, and Analytic Epidemiology, The University of Melbourne, Melbourne, Australia
| | - Sabina Rinaldi
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC), Lyon, France
| | - Isabelle Romieu
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC), Lyon, France
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition (DIfE), Nuthetal, Germany
| | - Antonia Trichopoulou
- Department of Hygiene, Epidemiology, and Medical Statistics, University of Athens Medical School, Athens, Greece. Hellenic Health Foundation, Athens, Greece
| | - Pagona Lagiou
- Department of Hygiene, Epidemiology, and Medical Statistics, University of Athens Medical School, Athens, Greece. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts. Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece
| | - Dimitrios Trichopoulos
- Hellenic Health Foundation, Athens, Greece. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts. Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece
| | - Giovanna Masala
- Molecular and Nutritional Epidemiology Unit, Cancer Research and Prevention Institute-ISPO, Florence, Italy
| | - Claudia Agnoli
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, "Civic-M.P.Arezzo" Hospital ASP, Ragusa, Italy
| | - Carlotta Sacerdote
- Center for Cancer Prevention (CPO-Piemonte), Torino, Italy. Human Genetic Foundation (HuGeF), Torino, Italy
| | - Amalia Mattiello
- Department of Clinical and Experimental Medicine, Federico II University, Naples, Italy
| | - Genevieve Buckland
- Unit of Nutrition, Environment, and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | | | - Esther Molina-Montes
- Escuela Andaluza de Salud Pública, Instituto de Investigación Biosanitario de Granada, Granada, Spain. Consortium for Biomedical Research in Epidemiology and Public Health (CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Pilar Amiano
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain. Basque Regional Health Department, Public Health Division of Gipuzkoa, San Sebastian, Spain
| | - José María Huerta Castaño
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain. Department of Epidemiology, Murcia Regional Health Authority, Murcia, Spain
| | - Aurelio Barricarte
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain. Navarre Public Health Institute, Pamplona, Spain
| | - H Bas Bueno-de-Mesquita
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands. Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, the Netherlands. School of Public Health, Imperial College, London, United Kingdom
| | - Evelyn M Monninkhof
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, the Netherlands
| | - N Charlotte Onland-Moret
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, the Netherlands
| | - Annika Idahl
- Department of Clinical Sciences, Obstetrics and Gynaecology, Faculty of Medicine, Umeå University, Umeå, Sweden
| | - Eva Lundin
- Department of Medical Biosciences, Pathology, Faculty of Medicine, Umeå University, Umeå, Sweden
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway. Department of Research, Cancer Registry of Norway, Oslo, Norway. Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden. Samfundet Folkhälsan, Helsinki, Finland
| | - Eiliv Lund
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Marit Waaseth
- Department of Pharmacy, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Kay-Tee Khaw
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Timothy J Key
- Cancer Epidemiology Unit, University of Oxford, Oxford, United Kingdom
| | - Ruth C Travis
- Cancer Epidemiology Unit, University of Oxford, Oxford, United Kingdom
| | - Marc J Gunter
- School of Public Health, Imperial College, London, United Kingdom
| | - Elio Riboli
- School of Public Health, Imperial College, London, United Kingdom
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
28
|
Abstract
AbstractStudies in both rodents and humans have made much progress in shedding light on how fluctuations in ovarian hormones can affect memory in women across the lifespan. Specifically, advances in neuroscience have identified multiple memory systems that are each mediated by different brain areas. Two memory systems used to navigate an environment are ‘place’ and ‘response’ memory. They are defined as either using an allocentric strategy: using a spatial or cognitive map of the surroundings, or an egocentric strategy: using habitual-turns/movements, respectively. Studies in neuroendocrinology have shown that estrogen levels can bias a female to use one memory system over another to solve a task, such that high estrogen levels are associated with using place memory and low levels with using response memory. Furthermore, recent advances in identifying and localizing estrogen receptors in the rodent brain are uncovering which brain regions are affected by estrogen and providing insight into how hormonal fluctuations during the menstrual cycle, pregnancy, and menopause might affect which memory system is facilitated or impaired in women at different life stages. These studies can help point the way to improving cognitive health in women.
Collapse
|
29
|
Meier-Abt F, Bentires-Alj M. How pregnancy at early age protects against breast cancer. Trends Mol Med 2013; 20:143-53. [PMID: 24355762 DOI: 10.1016/j.molmed.2013.11.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/17/2013] [Accepted: 11/18/2013] [Indexed: 12/11/2022]
Abstract
Pregnancy at an early age has a strong protective effect against breast cancer in humans and rodents. Postulated mechanisms underlying this phenomenon include alterations in the relative dynamics of hormone and growth factor-initiated cell fate-determining signaling pathways within the hierarchically organized mammary gland epithelium. Recent studies in epithelial cell subpopulations isolated from mouse and human mammary glands have shown that early pregnancy decreases the proportion of hormone receptor-positive cells and causes pronounced changes in gene expression as well as decreased proliferation in stem/progenitor cells. The changes include downregulation of Wnt and transforming growth factor β (TGFβ) signaling. These new findings highlight the importance of cell-cell interactions within the mammary gland epithelium in modulating cancer risk and provide potential targets for breast cancer prevention strategies.
Collapse
Affiliation(s)
- Fabienne Meier-Abt
- Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research (FMI), Maulbeerstrasse 66, CH-4058 Basel, Switzerland; Faculty of Science, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Mohamed Bentires-Alj
- Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research (FMI), Maulbeerstrasse 66, CH-4058 Basel, Switzerland.
| |
Collapse
|
30
|
Byrnes EM, Casey K, Carini LM, Bridges RS. Reproductive experience alters neural and behavioural responses to acute oestrogen receptor α activation. J Neuroendocrinol 2013; 25:1280-1289. [PMID: 24118285 PMCID: PMC4269101 DOI: 10.1111/jne.12113] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 09/04/2013] [Accepted: 10/09/2013] [Indexed: 01/25/2023]
Abstract
Reproductive experience (i.e. parturition and lactation) leads to persistent alterations in anxiety-like behaviour that are influenced by the oestrous cycle. We recently found that repeated administration of the selective oestrogen receptors (ER)α agonist propyl-pyrazole triol (PPT) results in anxiolytic-like behaviours on the elevated plus maze (EPM) in primiparous (but not nulliparous) female rats. The present study examined the effects of the acute administration of PPT on EPM behaviour in primiparous and aged-matched, nulliparous female rats. In addition, corticosterone secretion, corticotrophin-releasing hormone (CRH) gene expression and expression of the immediate early gene product Fos in the paraventricular nucleus (PVN) and amygdala were measured either after EPM testing or in home cage controls. Acute PPT administration significantly modified EPM behaviour as a function of reproductive experience, with nulliparous females tending toward increased anxiety-like behaviours and primiparous females tending toward decreased anxiety-like behaviours. In home cage controls, PPT increased corticosterone secretion in all females; however, both vehicle- and PPT-treated, primiparous females had reduced corticosterone levels compared to their nulliparous counterparts. Significant effects of PPT on CRH mRNA within the PVN were observed after the administration of PPT but only in primiparous females tested on the EPM. PPT also increased Fos expression within the PVN of EPM-exposed females; however, both vehicle- and PPT-treated primiparous females had reduced Fos expression compared to nulliparous females. In the amygdala, PPT increased Fos immunoreactivity in the central but not the medial or basolateral amygdala, although these effects were only observed in home cage females. Additionally, both vehicle- and PPT-treated home cage, primiparous females had increased Fos in the central nucleus of the amygdala compared to nulliparous controls. Overall, these data demonstrate that reproductive experience alters the behavioural response to acute ERα activation. Moreover, the findings suggest that central regulation of the hypothalamic-adrenal-pituitary axis is modified as a consequence of reproductive experience.
Collapse
Affiliation(s)
- E M Byrnes
- Tufts University, Cummings School of Veterinary Medicine, North Grafton, MA, USA
| | - K Casey
- Tufts University, Cummings School of Veterinary Medicine, North Grafton, MA, USA
| | - L M Carini
- Tufts University, Cummings School of Veterinary Medicine, North Grafton, MA, USA
| | - R S Bridges
- Tufts University, Cummings School of Veterinary Medicine, North Grafton, MA, USA
| |
Collapse
|
31
|
Ovarian hormone loss impairs excitatory synaptic transmission at hippocampal CA3-CA1 synapses. J Neurosci 2013; 33:16158-69. [PMID: 24107948 DOI: 10.1523/jneurosci.2001-13.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Premature and long-term ovarian hormone loss following ovariectomy (OVX) is associated with cognitive impairment. This condition is prevented by estradiol (E2) therapy when initiated shortly following OVX but not after substantial delay. To determine whether these clinical findings are correlated with changes in synaptic functions, we used adult OVX rats to evaluate the consequences of short-term (7-10 d, OVXControl) and long-term (∼5 months, OVXLT) ovarian hormone loss, as well as subsequent in vivo E2 treatment, on excitatory synaptic transmission at the hippocampal CA3-CA1 synapses important for learning and memory. The results show that ovarian hormone loss was associated with a marked decrease in synaptic strength. E2 treatment increased synaptic strength in OVXControl but not OVXLT rats, demonstrating a change in the efficacy for E2 5 months following OVX. E2 also had a more rapid effect: within minutes of bath application, E2 acutely increased synaptic strength in all groups except OVXLT rats that did not receive in vivo E2 treatment. E2's acute effect was mediated postsynaptically, and required Ca(2+) influx through the voltage-gated Ca(2+) channels. Despite E2's acute effect, synaptic strength of OVXLT rats remained significantly lower than that of OVXControl rats. Thus, changes in CA3-CA1 synaptic transmission associated with ovarian hormone loss cannot be fully reversed with delayed E2 treatment. Given that synaptic strength at CA3-CA1 synapses is related to the ability to learn hippocampus-dependent tasks, these findings provide additional insights for understanding cognitive impairment-associated long-term ovarian hormone loss and ineffectiveness for delayed E2 treatment to maintain cognitive functions.
Collapse
|
32
|
Evens AM, Advani R, Press OW, Lossos IS, Vose JM, Hernandez-Ilizaliturri FJ, Robinson BK, Otis S, Nadav Dagan L, Abdallah R, Kroll-Desrosiers A, Yarber JL, Sandoval J, Foyil K, Parker LM, Gordon LI, Blum KA, Flowers CR, Leonard JP, Habermann TM, Bartlett NL. Lymphoma Occurring During Pregnancy: Antenatal Therapy, Complications, and Maternal Survival in a Multicenter Analysis. J Clin Oncol 2013; 31:4132-9. [DOI: 10.1200/jco.2013.49.8220] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Lymphoma is the fourth most frequent cancer in pregnancy; however, current clinical practice is based largely on small series and case reports. Patients and Methods In a multicenter retrospective analysis, we examined treatment, complications, and outcomes for Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL) occurring during pregnancy. Results Among 90 patients (NHL, n = 50; HL, n = 40), median age was 30 years (range, 18 to 44 years) and median diagnosis occurred at 24 weeks gestation. Of patients with NHL, 52% had advanced-stage versus 25% of patients with HL (P = .01). Pregnancy was terminated in six patients. Among the other 84 patients, 28 (33%) had therapy deferred to postpartum; these patients were diagnosed at a median 30 weeks gestation. This compared with 56 patients (67%) who received antenatal therapy with median lymphoma diagnosis at 21 weeks (P < .001); 89% of these patients received combination chemotherapy. The most common preterm complication was induction of labor (33%). Gestation went to full term in 56% of patients with delivery occurring at a median of 37 weeks. There were no differences in maternal complications, perinatal events, or median infant birth weight based on deferred versus antenatal therapy. At 41 months, 3-year progression-free survival (PFS) and overall survival (OS) for NHL were 53% and 82%, respectively, and 85% and 97%, respectively, for HL. On univariate analysis for NHL, radiotherapy predicted inferior PFS, and increased lactate dehydrogenase and poor Eastern Cooperative Oncology Group performance status (ECOG PS) portended worse OS. For HL patients, nulliparous status and “B” symptoms predicted inferior PFS. Conclusion Standard (non-antimetabolite) combination chemotherapy administered past the first trimester, as early as 13 weeks gestation, was associated with few complications and expected maternal survival with lymphoma occurring during pregnancy.
Collapse
Affiliation(s)
- Andrew M. Evens
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Ranjana Advani
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Oliver W. Press
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Izidore S. Lossos
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Julie M. Vose
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Francisco J. Hernandez-Ilizaliturri
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Barrett K. Robinson
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Stavroula Otis
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Liat Nadav Dagan
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Ramsey Abdallah
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Aimee Kroll-Desrosiers
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Jessica L. Yarber
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Jose Sandoval
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Kelley Foyil
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Linda M. Parker
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Leo I. Gordon
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Kristie A. Blum
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Christopher R. Flowers
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - John P. Leonard
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Thomas M. Habermann
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| | - Nancy L. Bartlett
- Andrew M. Evens, Tufts University School of Medicine, Boston; Aimee Kroll-Desrosiers, University of Massachusetts Medical School, Worcester, MA; Ranjana Advani and Stavroula Otis, Stanford University Medical Center, Stanford, CA; Oliver W. Press and Linda M. Parker, Fred Hutchinson Cancer Research Center, Seattle, WA; Izidore S. Lossos, Liat Nadav Dagan, and Jose Sandoval, University of Miami School of Medicine, Miami, FL; Julie M. Vose, University of Nebraska Medical Center, Omaha, NE; Francisco J
| |
Collapse
|
33
|
Galea LAM, Wainwright SR, Roes MM, Duarte-Guterman P, Chow C, Hamson DK. Sex, hormones and neurogenesis in the hippocampus: hormonal modulation of neurogenesis and potential functional implications. J Neuroendocrinol 2013; 25:1039-61. [PMID: 23822747 DOI: 10.1111/jne.12070] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/23/2013] [Accepted: 06/29/2013] [Indexed: 12/12/2022]
Abstract
The hippocampus is an area of the brain that undergoes dramatic plasticity in response to experience and hormone exposure. The hippocampus retains the ability to produce new neurones in most mammalian species and is a structure that is targeted in a number of neurodegenerative and neuropsychiatric diseases, many of which are influenced by both sex and sex hormone exposure. Intriguingly, gonadal and adrenal hormones affect the structure and function of the hippocampus differently in males and females. Adult neurogenesis in the hippocampus is regulated by both gonadal and adrenal hormones in a sex- and experience-dependent way. Sex differences in the effects of steroid hormones to modulate hippocampal plasticity should not be completely unexpected because the physiology of males and females is different, with the most notable difference being that females gestate and nurse the offspring. Furthermore, reproductive experience (i.e. pregnancy and mothering) results in permanent changes to the maternal brain, including the hippocampus. This review outlines the ability of gonadal and stress hormones to modulate multiple aspects of neurogenesis (cell proliferation and cell survival) in both male and female rodents. The function of adult neurogenesis in the hippocampus is linked to spatial memory and depression, and the present review provides early evidence of the functional links between the hormonal modulation of neurogenesis that may contribute to the regulation of cognition and stress.
Collapse
Affiliation(s)
- L A M Galea
- Department of Psychology, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Carvalho-Freitas MIR, Anselmo-Franci JA, Palermo-Neto J, Felicio LF. Prior reproductive experience alters prolactin-induced macrophage responses in pregnant rats. J Reprod Immunol 2013; 99:54-61. [DOI: 10.1016/j.jri.2013.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 03/25/2013] [Accepted: 03/26/2013] [Indexed: 10/26/2022]
|
35
|
Workman JL, Crozier T, Lieblich SE, Galea LAM. Reproductive experience does not persistently alter prefrontal cortical-dependent learning but does alter strategy use dependent on estrous phase. Horm Behav 2013; 64:439-47. [PMID: 23827889 DOI: 10.1016/j.yhbeh.2013.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 06/21/2013] [Accepted: 06/23/2013] [Indexed: 11/16/2022]
Abstract
Reproductive experiences in females comprise substantial hormonal and experiential changes and can exert long lasting changes in cognitive function, stress physiology, and brain plasticity. The goal of this research was to determine whether prior reproductive experience could alter a prefrontal-cortical dependent form of learning (strategy set shifting) in an operant box. In this study, female Sprague-Dawley rats were mated and mothered once or twice to produce either primiparous or biparous dams, respectively. Age-matched nulliparous controls (reproductively-naïve females with no exposure to pup cues) were also used. Maternal behaviors were also assessed to determine whether these factors would predict cognitive flexibility. For strategy set shifting, rats were trained in a visual-cue discrimination task on the first day and on the following day, were required to switch to a response strategy to obtain a reward. We also investigated a simpler form of behavioral flexibility (reversal learning) in which rats were trained to press a lever on one side of the box the first day, and on the following day, were required to press the opposite lever to obtain a reward. Estrous phase was determined daily after testing. Neither parity nor estrous phase altered total errors or trials to reach criterion in either the set-shifting or reversal-learning tasks, suggesting that PFC-dependent cognitive performance remains largely stable after 1 or 2 reproductive experiences. However, parity and estrous phase interacted to alter the frequency of particular error types, with biparous rats in estrus committing more perseverative but fewer regressive errors during the set-shifting task. This suggests that parity and estrous phase interfere with the ability to disengage from a previously used, but no longer relevant strategy. These data also suggest that parity alters the behavioral sensitivity to ovarian hormones without changing overall performance.
Collapse
Affiliation(s)
- Joanna L Workman
- Department of Psychology, University of British Columbia, Canada
| | | | | | | |
Collapse
|
36
|
Effect of multiparity on morphometry and oestrogen receptor expression of pelvic and perineal striated muscles in rabbits: is serum oestradiol relevant? Eur J Obstet Gynecol Reprod Biol 2013; 169:113-20. [PMID: 23664379 DOI: 10.1016/j.ejogrb.2013.03.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 01/11/2013] [Accepted: 03/25/2013] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine changes in morphometry and expression of oestrogen receptors (OR) in the pubococcygeus and bulbospongiosus muscles, and the concentration of serum oestradiol associated with multiparity. STUDY DESIGN Twelve chinchilla-breed female rabbits were divided into multiparas who had undergone four consecutive deliveries and age-matched virgin nulliparas. Pubococcygeus and bulbospongiosus muscles were surgically removed from each rabbit and processed histologically. Fibre cross-sectional area, number of peripheral nuclei, and expression of ORα and ORβ were measured for each muscle. Serum samples were obtained and the concentration of serum oestradiol was quantified. RESULTS Multiparity increased (p ≤ 0.05) fibre cross-sectional area and the number of peripheral nuclei per fibre in pubococcygeus muscle, but not in bulbospongiosus muscle. Expression of both ORα and ORβ was high (p ≤ 0.05) in both muscles from multiparous rabbits. A rise in serum oestradiol was measured at the end of the second pregnancy, which was absent (p ≤ 0.05) at the end of the fourth pregnancy. The concentration of serum oestradiol was similar (p > 0.05) in nulliparous and multiparous rabbits. CONCLUSIONS Multiparity caused morphometric changes in pubococcygeus muscle but not in bulbospongiosus muscle. As OR expression was high for both muscles, some properties related to fibre composition or muscle physiology could be affected. The finding that serum oestradiol was not elevated at the end of the fourth pregnancy could be related to changes in pelvic and perineal muscles associated with multiparity.
Collapse
|
37
|
Hussain D, Hoehne A, Woodside B, Brake WG. Reproductive experience modifies the effects of estradiol on learning and memory bias in female rats. Horm Behav 2013. [PMID: 23195751 DOI: 10.1016/j.yhbeh.2012.11.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Previous studies have shown that estrogen affects whether a hippocampus-mediated place (allocentric) or a striatum-mediated response (egocentric) memory system is employed by female rats when searching for a food reward in a maze. Because it has been suggested that reproductive experience alters some of the responses to E in the brain, two experiments were carried out to investigate whether reproductive experience would also alter the effect of E on place and response learning. In experiment 1, 152 ovariectomized nulliparous (n=77; no reproductive experience) and primiparous (n=74; having had and raised one litter of pups) Wistar rats were trained on an ambiguous t-maze task and tested for memory system bias. In experiment 2, 35 ovariectomized nulliparous (n=16) and primiparous (n=19) Wistar rats were trained on place and response plus-maze tasks. All rats were exposed to no, chronic low or chronic low with pulsatile high 17β-estradiol (E2) replacement. Congruent with previous findings, low E2 nulliparous rats showed predominant use of response memory and faster response learning, whereas high E2 nulliparous rats showed a trend towards predominant place memory use. Interestingly, the facilitatory effect of low E2 on response task learning and memory seen in nulliparous rats was not observed in low E2 primiparous rats in either experiment. In conclusion, E2 levels do dictate the rate at which female rats learn a response task and utilize response memory, but only in those with no reproductive experience.
Collapse
Affiliation(s)
- Dema Hussain
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, 7141 Sherbrooke St. W, Room SP-244, Montreal, Canada H4B 1R6.
| | | | | | | |
Collapse
|
38
|
Babb JA, Masini CV, Day HEW, Campeau S. Sex differences in activated corticotropin-releasing factor neurons within stress-related neurocircuitry and hypothalamic-pituitary-adrenocortical axis hormones following restraint in rats. Neuroscience 2013; 234:40-52. [PMID: 23305762 DOI: 10.1016/j.neuroscience.2012.12.051] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 12/20/2012] [Accepted: 12/28/2012] [Indexed: 02/06/2023]
Abstract
Women may be more vulnerable to certain stress-related psychiatric illnesses than men due to differences in hypothalamic-pituitary-adrenocortical (HPA) axis function. To investigate potential sex differences in forebrain regions associated with HPA axis activation in rats, these experiments utilized acute exposure to a psychological stressor. Male and female rats in various stages of the estrous cycle were exposed to 30min of restraint, producing a robust HPA axis hormonal response in all animals, the magnitude of which was significantly higher in female rats. Although both male and female animals displayed equivalent c-fos expression in many brain regions known to be involved in the detection of threatening stimuli, three regions had significantly higher expression in females: the paraventricular nucleus of the hypothalamus (PVN), the anteroventral division of the bed nucleus of the stria terminalis (BSTav), and the medial preoptic area (MPOA). Dual fluorescence in situ hybridization analysis of neurons containing c-fos and corticotropin-releasing factor (CRF) mRNA in these regions revealed significantly more c-fos and CRF single-labeled neurons, as well as significantly more double-labeled neurons in females. Surprisingly, there was no effect of the estrous cycle on any measure analyzed, and an additional experiment revealed no demonstrable effect of estradiol replacement following ovariectomy on HPA axis hormone induction following stress. Taken together, these data suggest sex differences in HPA axis activation in response to perceived threat may be influenced by specific populations of CRF neurons in key stress-related brain regions, the BSTav, MPOA, and PVN, which may be independent of circulating sex steroids.
Collapse
Affiliation(s)
- J A Babb
- Department of Psychology and Neuroscience, University of Colorado at Boulder, USA
| | | | | | | |
Collapse
|
39
|
Keiler AM, Bernhardt R, Scharnweber D, Jarry H, Vollmer G, Zierau O. Comparison of estrogenic responses in bone and uterus depending on the parity status in Lewis rats. J Steroid Biochem Mol Biol 2013; 133:101-9. [PMID: 23032373 DOI: 10.1016/j.jsbmb.2012.09.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 09/04/2012] [Accepted: 09/24/2012] [Indexed: 10/27/2022]
Abstract
The reproductive transition of women through peri- to postmenopause is characterized by changes in steroid hormone levels due to the cessation of the ovarian function. Beside several complaints associated with these hormonal changes, the deterioration of the trabecular bone micro-architecture and the loss of skeletal mass can cause osteoporosis. At this life stage, women often have a reproductive history of one to several pregnancies. The ovariectomized skeletally mature rat (>10 months old) is one of the most commonly used animal models for postmenopausal osteoporosis research. Despite the fact that mammals can undergo up to several reproductive cycles (primi-/pluriparous), nulliparous animals are often used and the question whether changes in the hormonal milieu subsequently affect the skeleton and influence the outcome of intervention studies is often neglected in study designs. Therefore, the aim of the present study was to compare the estrogen responsiveness of nulliparous and pluriparous rats. For this purpose, one year old virgin or retired breeder Lewis rats were either sham operated or ovariectomized, whereas half of the ovariectomized animals received subcutaneous 17β-estradiol pellets eight weeks after surgery. After another four weeks, the effects on the uterus were determined by expression analysis of estrogen-dependently regulated steroid receptor genes and well-established marker genes. Moreover, trabecular bone parameters in the tibia were analyzed by micro-computed tomography (μCT). Parity-dependency in estrogen responsiveness was observed with respect to the achieved serum E2 levels in response to similar E2 treatment. This led to differences both on the uterus wet weight and on the expression level of uterine target genes. In addition, a reversal of the ovariectomy-induced changes of the bone architecture after 17β-estradiol substitution was only observed among the nulliparous. In conclusion, the observations of this study support parity-dependent differences in the responses to estrogenic compounds in the uterus and the bone of rats. These results indicate that the parity-status has an impact on the outcome of studies aiming at the investigation of estrogenic effects of compounds potentially used in hormone replacement and thus, this should be taken into consideration for further studies and particularly for the discussion of data obtained with the preclinical ovariectomized rat animal model.
Collapse
Affiliation(s)
- Annekathrin Martina Keiler
- Institute of Zoology, Molecular Cell Physiology and Endocrinology, Technische Universität Dresden, 01062 Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
In the rat, the change from a virgin/nulliparous female to the maternal animal takes place at many levels. A subtle developmental wave washes over the female nervous system and transforms her from largely self-centred to offspring-directed, from personal care and protection to care of genetically-related offspring, from indifference to ardour. Such change is preceded by substantial and apparently permanent neural alterations, the depth of which results in the maternal brain, and is the basis of the present review. The neuroplasticity of pregnancy, inherent to the female brain and, we believe, representative of the full expression of the female nervous system's capacity, is a result of significant hormonal and other neurochemical actions. It results in the striking brain changes that are associated with, and necessary for, successful reproduction. We discuss some of these changes and their ramifications. Collectively, they represent the culmination of mammalian evolution and have led to the development of the social brain characteristic of higher orders of mammal, including the human. We also examine different facets of the maternal brain, beginning with a review of the genes involved in maternal behaviour, and in the subsequent 'expression' of the maternal brain. We next discuss olfaction and the manner in which this major sense draws from the rich sensory milieu of the mother to regulate and support maternal behaviour. Last, we discuss the 'whys' of maternal behaviour, a theoretical foray into the reasons for such substantial maternal brain alterations. We focus on the male's potential role as the raison d'etre for the manifest alterations in his mate's brain. In the end, it is clear that the female brain undergoes a significant reorganisation en route to motherhood, the results of which are deep and enduring.
Collapse
Affiliation(s)
- C H Kinsley
- Department of Psychology-Center for Neuroscience, University of Richmond, Richmond, VA 23173, USA.
| | | |
Collapse
|
41
|
Franssen CL, Bardi M, Shea EA, Hampton JE, Franssen RA, Kinsley CH, Lambert KG. Fatherhood alters behavioural and neural responsiveness in a spatial task. J Neuroendocrinol 2011; 23:1177-87. [PMID: 21933288 DOI: 10.1111/j.1365-2826.2011.02225.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The hormones and experiences of pregnancy, parturition and lactation have been shown to dramatically remodel the female rat's hippocampus, potentially enhancing behaviours critical for meeting the increased demands of motherhood. Previous work in our laboratory has also suggested that pup exposure, apart from pregnancy and lactation, constitutes an important influence on ancillary maternal behaviour (e.g. foraging behaviour). In the present study, we press the parental model further by examining the effect of pup exposure on the hippocampus of males from a biparental mouse species, the California mice (Peromyscus californicus). Males were either Fathers (i.e. first-time fathers housed with a female from mating until 7 days after parturition), pup-exposed virgins (PEV; i.e. sexually naïve males briefly exposed to pups daily for 7 days) or Virgins (i.e. never exposed to females or pups). A dry-land maze (DLM), as used for assessing spatial learning, was employed to determine the foraging abilities of the males. The results indicated that, on the most challenging day of testing (i.e. acquisition day), California mouse Fathers demonstrated superior memory for the task compared to PEVs and Virgins. In addition to the behavioural data, significantly more fos-immunoreactivity was observed in the CA1, CA3 and dentate gyrus regions of the hippocampi of Fathers than PEVs or Virgins in response to the probe trial. Additionally, a trend for altered performance on the DLM was observed in the PEVs on the last day of testing, which was accompanied by the highest levels of nestin-immunoreactivity, an indicant of neuroplasticity, of the three groups. In summary, these data suggest that, in accordance with previous observations of maternal rats, the paternal brain is similarly influenced by parental experience, as demonstrated by accompanying modifications to relevant neurobiological and behavioural responses.
Collapse
Affiliation(s)
- C L Franssen
- Department of Psychology, Randolph-Macon College, Ashland, VA 23005, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Royal CR, Ma H, Walker R, White RE. Estrogen signaling in microvascular arteries: parturition reduces vasodilation by reducing 17β-estradiol and nNOS. Steroids 2011; 76:991-7. [PMID: 21458475 PMCID: PMC3139781 DOI: 10.1016/j.steroids.2011.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 03/18/2011] [Accepted: 03/24/2011] [Indexed: 12/28/2022]
Abstract
Few studies have examined the potential effects of childbirth on the responses of the female vasculature--especially the resistance microvasculature of non-reproductive tissues. In the present study we have investigated the response of mesenteric microvascular resistance vessels to estrogen (E2), an important vasoactive hormone. Vessels were obtained from either nulliparous or postpartum female Sprague-Dawley rats, and isometric tension studies were performed. We found that E2 induced a concentration-dependent, endothelium-independent relaxation of microvessels precontracted with 10(-5) M phenylephrine; however, E2-induced relaxation was reduced by nearly half in vessels from postpartum animals compared to nulliparous controls. Inhibiting nitric oxide synthase activity with 10(-4) M L-NMMA or L-NPA (which exhibits selectivity for type 1 or nNOS) attenuated the relaxation effect of E2 on arteries from nulliparous animals. In contrast, L-NPA had little effect on arteries from postpartum animals, suggesting a reduced influence of nNOS after parturition. Moreover, expression of nNOS protein in microvessels was decreased 39% in the postpartum state compared to arteries from nulliparous animals. We propose that the impaired E2-induced relaxation response of microvessels from postpartum animals reflects a downregulation of NO production due to lower nNOS expressed in vascular smooth muscle cells. We measured a 73% decrease in serum E2 levels in the postpartum state compared to nulliparous animals. Because E2 has been shown to increase nNOS protein expression, we propose that lower E2 levels after parturition decrease expression of nNOS, leading to a reduced vasodilatory capacity of resistance microvessels.
Collapse
Affiliation(s)
- Crista R. Royal
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA 30912
| | - Handong Ma
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA 30912
| | - Richard Walker
- Department of Biostatistics, Georgia Health Sciences University, Augusta, GA 30912
| | - Richard E. White
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA 30912
| |
Collapse
|
43
|
Martínez-Gómez M, Mendoza-Martínez G, Corona-Quintanilla DL, Fajardo V, Rodríguez-Antolín J, Castelán F. Multiparity causes uncoordinated activity of pelvic- and perineal-striated muscles and urodynamic changes in rabbits. Reprod Sci 2011; 18:1246-52. [PMID: 21701040 DOI: 10.1177/1933719111411728] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Temporal and coordinated activation of pelvic- (pubococcygeous) and perineal- (bulbospongiosus and ischiocavernosus) striated muscles occurs during micturition in female rabbits. We have hypothesized that the coordinated activation of pelvic and perineal muscles is modified during the micturition of young multiparous rabbits. Young virgin and multiparous female chinchilla rabbits were used to simultaneously record cystometrograms and electromyograms of the pubococcygeous, ischocavernosus, and bulbospongiosus muscles. Bladder function was assessed using standard urodynamic variables. The temporal coordination of pelvic- and perineal-striated muscle activity was changed in multiparous rabbits. The cystometrogram recordings were different than those obtained from virgin rabbits, as seen in alterations of the threshold volume, the residual volume, the voiding duration, and the maximum pressure. In rabbits, we find that multiparity causes uncoordinated activity of pubococcygeous, ischiocavernosus, and bulbospongiosus muscles and modifies the urodynamics.
Collapse
Affiliation(s)
- Margarita Martínez-Gómez
- Depto. de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México DF, México.
| | | | | | | | | | | |
Collapse
|
44
|
Lambert KG, Franssen CL, Bardi M, Hampton JE, Hainley L, Karsner S, Tu EB, Hyer MM, Crockett A, Baranova A, Ferguson T, Ferguson T, Kinsley CH. Characteristic neurobiological patterns differentiate paternal responsiveness in two Peromyscus species. BRAIN, BEHAVIOR AND EVOLUTION 2011; 77:159-75. [PMID: 21546770 DOI: 10.1159/000326054] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 02/14/2011] [Indexed: 11/19/2022]
Abstract
Rodent paternal models provide unique opportunities to investigate the emergence of affiliative social behavior in mammals. Using biparental and uniparental Peromyscus species (californicus and maniculatus, respectively) we assessed paternal responsiveness by exposing males to biological offspring, unrelated conspecific pups, or familiar brothers following a 24-hour separation. The putative paternal circuit we investigated included brain areas involved in fear/anxiety [cingulate cortex (Cg), medial amygdala (MeA), paraventricular nucleus of the hypothalamus (PVN), and lateral septum (LS)], parental motivation [medial preoptic area (MPOA)], learning/behavioral plasticity (hippocampus), olfaction [pyriform cortex (PC)], and social rewards (nucleus accumbens). Paternal experience in californicus males reduced fos immunoreactivity (ir) in several fear/anxiety areas; additionally, all californicus groups exhibited decreased fos-ir in the PC. Enhanced arginine vasopressin (AVP) and oxytocin (OT)-ir cell bodies and fibers, as well as increased neuronal restructuring in the hippocampus, were also observed in californicus mice. Multidimensional scaling analyses revealed distinct brain activation profiles differentiating californicus biological fathers, pup-exposed virgins, and pup-naïve virgins. Specifically, associations among MPOA fos, CA1 fos, dentate gyrus GFAP, CA2 nestin-, and PVN OT-ir characterized biological fathers; LS fos-, Cg fos-, and AVP-ir characterized pup-exposed virgins, and PC-, PVN-, and MeA fos-ir characterized pup-naïve virgins. Thus, whereas fear/anxiety areas characterized pup-naïve males, neurobiological factors involved in more diverse functions such as learning, motivation, and nurturing responses characterized fatherhood in biparental californicus mice. Less distinct paternal-dependent activation patterns were observed in uniparental maniculatus mice. These data suggest that dual neurobiological circuits, leading to the inhibition of social-dependent anxiety as well as the activation of affiliative responses, characterize the transition from nonpaternal to paternal status in californicus mice.
Collapse
Affiliation(s)
- Kelly G Lambert
- Department of Psychology, Randolph-Macon College, Ashland, Va., USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sjoeholm A, Bridges RS, Grattan DR, Anderson GM. Region-, neuron-, and signaling pathway-specific increases in prolactin responsiveness in reproductively experienced female rats. Endocrinology 2011; 152:1979-88. [PMID: 21363933 PMCID: PMC3075931 DOI: 10.1210/en.2010-1220] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pregnancy and lactation cause long-lasting enhancements in maternal behavior and other physiological functions, along with increased hypothalamic prolactin receptor expression. To directly test whether reproductive experience increases prolactin responsiveness in the arcuate, paraventricular, and supraoptic nuclei and the medial preoptic area, female rats experienced a full pregnancy and lactation or remained as age-matched virgin controls. At 5 wk after weaning, rats received 2.5, 100, or 4000 ng ovine prolactin or vehicle intracerebroventricularly. The brains underwent immunohistochemistry for the phosphorylated forms of signal transducer and activator of transcription 5 (pSTAT5) or ERK1/2 (pERK1/2). There was a marked increase in pSTAT5 and pERK1/2 in response to prolactin in the regions examined in both virgin and primiparous rats. Primiparous rats exhibited approximately double the number of prolactin-induced pSTAT5-immunoreactive cells as virgins, this effect being most apparent at the higher prolactin doses in the medial preoptic area and paraventricular and supraoptic nuclei and at the lowest prolactin dose in the arcuate nucleus. Dual-label immunohistochemistry showed that arcuate kisspeptin (but not oxytocin or dopamine) neurons displayed increased sensitivity to prolactin in reproductively experienced animals; these neurons may contribute to the reduction in prolactin concentration observed after reproductive experience. There was no effect of reproductive experience on prolactin-induced pERK1/2, indicating a selective effect on the STAT5 pathway. These data show that STAT5 responsiveness to prolactin is enhanced by reproductive experience in multiple hypothalamic regions. The findings may have significant implications for understanding postpartum disorders affecting maternal care and other prolactin-associated pathologies.
Collapse
Affiliation(s)
- Annika Sjoeholm
- Centre for Neuroendocrinology and Department of Anatomy and Structural Biology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | | | | | | |
Collapse
|
46
|
Bridges RS, Scanlan VF, Lee JO, Byrnes EM. Reproductive experience alters prolactin receptor expression in mammary and hepatic tissues in female rats. Biol Reprod 2011; 85:340-6. [PMID: 21508351 DOI: 10.1095/biolreprod.111.091918] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Recent studies have reported that reproductive experience in female rats alters prolactin (PRL) receptor gene expression in the brain as well as neural sensitivity to PRL. Given PRL's actions in nonneural tissues, that is, mammary tissue and liver, it was asked whether reproductive experience may also alter prolactin receptor (Prlr) gene expression in these tissues. Groups of age-matched female rats were generated with varying reproductive histories. Separate groups of primiparous (first lactation) and multiparous (second lactation) had mammary tissue and liver samples collected on Day 3 or 10 of lactation. A fifth group raised one litter to weaning and then resumed estrous cyclicity. This group and a final group of age-matched, virgin controls were killed on diestrus. Tissue was processed by quantitative PCR for expression rates of the long and short forms of Prlr mRNA as well as casein beta mRNA (mammary tissue only). Western blots were performed to quantify receptor protein content. Multiple lactations as well as lactation itself resulted in alterations in Prlr expression. Prlr gene expression in mammary tissue was increased in primiparous mothers compared with that in multiparous dams, whereas in the liver, Prlr expression was reduced during an initial lactation. In contrast, PRLR protein levels declined during lactation in mammary, but not hepatic, tissues. Overall, the results demonstrate that the prolactin receptor system is altered in nonneural tissues as a result of the female's reproductive history. The findings are discussed in the context of milk and bile production and PRL's possible role in breast cancer.
Collapse
Affiliation(s)
- Robert S Bridges
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA.
| | | | | | | |
Collapse
|
47
|
Carvalho-Freitas MIR, Anselmo-Franci JA, Maiorka PC, Palermo-Neto J, Felicio LF. Prolactin differentially modulates the macrophage activity of lactating rats: possible role of reproductive experience. J Reprod Immunol 2011; 89:38-45. [DOI: 10.1016/j.jri.2010.12.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 11/17/2010] [Accepted: 12/02/2010] [Indexed: 11/26/2022]
|
48
|
Walf AA, Paris JJ, Rhodes ME, Simpkins JW, Frye CA. Divergent mechanisms for trophic actions of estrogens in the brain and peripheral tissues. Brain Res 2011; 1379:119-36. [PMID: 21130078 PMCID: PMC3103067 DOI: 10.1016/j.brainres.2010.11.081] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 11/23/2010] [Accepted: 11/23/2010] [Indexed: 01/08/2023]
Abstract
17β-estradiol (E(2)) can enhance reproductive, cognitive, and affective functions; however, the mechanisms by which E(2) has these effects need to be better understood. Pleiotrophic effects of E(2) can occur via traditional and novel actions at various forms of estrogen receptors (ERs). In the central nervous system, trophic effects of E(2) may be related to beneficial effects of hormone replacement therapy (HRT). However, in peripheral reproductive tissues, E(2)'s capacity to evoke growth can increase risk of cancers. This review focuses on investigations aimed at elucidating divergent mechanisms of steroids to promote trophic effects in the brain, independent of effects on peripheral reproductive tissues. First, actions of estrogens via ERα or ERβ for peripheral growth (carcinogen-induced tumors, uterine growth) and hippocampus-dependent behaviors (affect, cognition) are described. Second, factors that influence these effects of estrogens are described (e.g. experience, timing/critical windows, non-ER mechanisms). Third, effects of estrogens at ERβ related to actions of progestogens, such as 5α-pregnan-3α-ol-20-one (3α,5α-THP) are described. In summary, effects of E(2) may occur via multiple mechanisms, which may underlie favorable effects in the brain with minimal peripheral trophic effects.
Collapse
Affiliation(s)
- Alicia A. Walf
- Life Sciences Research, University at Albany, Albany, NY 12222, USA
| | - Jason J. Paris
- Department of Psychology, University at Albany, Albany, NY 12222, USA
| | | | - James W. Simpkins
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Cheryl A. Frye
- Department of Psychology, University at Albany, Albany, NY 12222, USA
- Department of Biological Sciences, University at Albany, Albany, NY 12222, USA
- Center for Neuroscience Research, University at Albany, Albany, NY 12222, USA
- Life Sciences Research, University at Albany, Albany, NY 12222, USA
| |
Collapse
|
49
|
Wexler A, Sand T, Elsås TB. Macular thickness measurements in healthy Norwegian volunteers: an optical coherence tomography study. BMC Ophthalmol 2010; 10:13. [PMID: 20465801 PMCID: PMC2885325 DOI: 10.1186/1471-2415-10-13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 05/13/2010] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ethnic, intersubject, interoperator and intermachine differences in measured macular thickness seem to exist. Our purpose was to collect normative macular thickness data in Norwegians and to evaluate the association between macular thickness and age, gender, parity, and contraception status. METHODS Retinal thickness was measured by Stratus Optical Coherence Tomography in healthy subjects. Mean macular thickness (MMT) was analyzed by repeated measures ANOVA with three dependent regional MMT-variables for interaction with age, gender, parity and oral contraception use. Exploratory correlation with age by the Pearson correlation test, both before and after stratification by gender was performed. Differences in MMT between older and younger subjects, between oral contraception users and non-users, as well as parous and nulliparous women were studied by post-hoc Student's t-tests. RESULTS Central MMT in Norwegians was similar to values earlier reported in whites. MMT in central areas of 1 and 2.25 mm in diameter were higher in males than in females. In younger subjects (< or =43 years) differences in MMT between genders were larger than in the mixed age group, whereas in older subjects (>43 years) the small differences did not reach the set significance level. No differences were found in minimal foveolar thickness (MMFT) between the genders in any age group.Mean foveal thickness (1 mm in diameter) was positively associated with age in females (r = 0.28, p = 0.03). MMFT was positively associated with age in all groups and reached significance both in females and in mixed gender group (r = 0.20, p = 0.041 and r = 0.26, p = 0.044 respectively).Mean foveal thickness and MMFT were significantly higher in parous than in nulliparous women, and age-adjusted ANOVA for MMFT revealed a borderline effect of parity. CONCLUSIONS Age and gender should be taken into consideration when establishing normal ranges for MMT in younger subjects. The gender difference in retinal thickness in young, but not older adults suggests a gonadal hormonal influence. The possible association between parity and retinal structure and its clinical relevance, should be studied further.
Collapse
Affiliation(s)
- Alexandra Wexler
- Department of Ophthalmology, St. Olavs University Hospital, Trondheim, Norway.
| | | | | |
Collapse
|
50
|
Pawluski JL, Barakauskas VE, Galea LAM. Pregnancy decreases oestrogen receptor alpha expression and pyknosis, but not cell proliferation or survival, in the hippocampus. J Neuroendocrinol 2010; 22:248-57. [PMID: 20136685 DOI: 10.1111/j.1365-2826.2010.01960.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Motherhood differentially affects learning and memory performance and this effect depends on reproductive experience. In turn, evidence suggests that the effects of oestradiol on learning and memory are mediated through binding to oestrogen receptors in the hippocampus and that this is related to hippocampal neurogenesis. The present study investigated the effect of pregnancy and reproductive experience on ERalpha expression throughout the hippocampus, as well as cell proliferation, new cell survival and cell death (as measured by pyknotic cells) in the granule cell layer of the hippocampus. Three groups of female Sprague-Dawley rats were used: virgin, primigravid and multigravid. All rats were injected with 5-bromo-2-deoxyuridine (BrdU; 200 mg/kg) on the afternoon of impregnation and at matched time-points in virgins. Rats were perfused either during early pregnancy (gestation day 1) or late pregnancy (gestation day 21) after BrdU injection. The results obtained show that, during late pregnancy, females, whether first or second pregnancy, have fewer ERalpha-positive cells in the CA3 region of the dorsal hippocampus than virgin females. In addition during early pregnancy, females have significantly fewer pyknotic cells in the granule cell layer than virgin females. There were no other differences between groups in the number of ERalpha-positive, BrdU-positive or pyknotic cells. Future studies will aim to investigate the mechanisms and consequences of the alteration in ERalpha expression in the hippocampus during late pregnancy, as well as the possible changes in ERbeta expression at this time.
Collapse
Affiliation(s)
- J L Pawluski
- Program in Neuroscience, Department of Psychology and Brain Research Centre, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|