1
|
Curi TZ, Passoni MT, Tolouei SEL, de Araújo Ramos AT, de Almeida SCF, Romano RM, de Oliveira JM, Dalsenter PR, Martino-Andrade AJ. In Utero and Lactational Exposure to an Environmentally Relevant Mixture of Phthalates Alters Hypothalamic Gene Expression and Sexual Preference in Rats. ENVIRONMENTAL TOXICOLOGY 2025; 40:54-65. [PMID: 39248502 DOI: 10.1002/tox.24414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
Several phthalates, mainly used as plasticizers, are known for their adverse effects on the male genital system. Previously, we demonstrated that an environmentally relevant mixture of six antiandrogenic phthalates (PMix), derived from a biomonitoring study in pregnant Brazilian women, was able to disrupt the reproductive development in male rats. Experimental groups (control, 0.1, 0.5, and 500 mg PMix/kg/day) were established starting from the extrapolated human dose (0.1 mg/kg/day), followed by doses 5 times and 5000 times higher. Pregnant rats received daily oral gavage administration of either vehicle (control) or PMix from gestational day 13 to postnatal day 10. Here, we examined male and female offspring regarding changes in gene expression of key reproductive factors in the hypothalamus and pituitary gland at adulthood and conducted a battery of behavioral tests in males, including partner preference, sexual behavior, and male attractiveness tests. PMix induced some changes in mating-related behavior in males, as demonstrated by the absence of preference for females against males and a higher number of penetrations up to ejaculation in the 0.5 dose group. PMix decreased Esr2 expression in the male hypothalamus across all three doses, and in females at mid and high doses in both the hypothalamus and pituitary. In male hypothalamus, we also observed decreased Kiss1 transcripts in these groups and a reduction in AR at the 0.5 dose group. In summary, our results provide further evidence that phthalates in a mixture, even at low doses, may exert cumulative effects on the structures underlying sexual behavior, which seems to be more sensitive than reproductive endpoints for the same experimental design.
Collapse
Affiliation(s)
- Tatiana Zauer Curi
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Marcella Tapias Passoni
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Sara Emilia Lima Tolouei
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Anderson Tadeu de Araújo Ramos
- Animal Endocrine and Reproductive Physiology Laboratory, Department of Physiology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Samara Christina França de Almeida
- Animal Endocrine and Reproductive Physiology Laboratory, Department of Physiology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Renata Marino Romano
- Reproductive Toxicology Laboratory, Department of Pharmacy, State University of Centro-Oeste, Guarapuava, Brazil
| | - Jeane Maria de Oliveira
- Reproductive Toxicology Laboratory, Department of Pharmacy, State University of Centro-Oeste, Guarapuava, Brazil
| | - Paulo Roberto Dalsenter
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Anderson Joel Martino-Andrade
- Reproductive Toxicology Laboratory, Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, Brazil
- Animal Endocrine and Reproductive Physiology Laboratory, Department of Physiology, Federal University of Paraná (UFPR), Curitiba, Brazil
| |
Collapse
|
2
|
Pfaus JG, García-Juárez M, Ordóñez RD, Tecamachaltzi-Silvarán MB, Lucio RA, González-Flores O. Cellular and molecular mechanisms of action of ovarian steroid hormones II: Regulation of sexual behavior in female rodents. Neurosci Biobehav Rev 2025; 168:105946. [PMID: 39571668 DOI: 10.1016/j.neubiorev.2024.105946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/25/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024]
Abstract
Female sexual behaviors in rodents (lordosis and appetitive or "proceptive" behaviors) are induced through a genomic mechanism by the sequential actions of estradiol (E2) and progesterone (P), or E2 and testosterone (T) at their respective receptors. However, non-steroidal agents, such as gonadotropin-releasing hormone (GnRH), Prostaglandin E2 (PGE2), noradrenaline, dopamine, oxytocin, α-melanocyte stimulating hormone, nitric oxide, leptin, apelin, and others, facilitate different aspects of female sexual behavior through their cellular and intracellular effects at the membrane and genomic levels in ovariectomized rats primed with E2. These neurotransmitters often act as intermediaries of E2 and P (or T). The classical model of steroid hormone action through intracellular receptor binding has been complemented by an alternative scenario wherein the steroid functions as a transcription factor after binding the receptor protein to DNA. Another possible mechanism occurs through the activation of second messenger systems (cyclic AMP, cyclic GMP, calcium), which subsequently initiate phosphorylation events via diverse kinase systems (protein kinases A, G, or C). These kinases target the progesterone receptor (PR) or associated effector proteins that connect the PR to the trans-activation machinery. This may also happen to the androgen receptor (AR). In addition, other cellular mechanisms could be involved since the chemical structure of these non-steroidal agents causes a change in their lipophobicity that prevents them from penetrating the cell and exerting direct transcriptional effects; however, they can exert effects on different components of the cell membrane activating a cross-talk between the cell membrane and the regulation of the transcriptional mechanisms.
Collapse
Affiliation(s)
- James G Pfaus
- Center for Sexual Health and Intervention, Czech National Institute of Mental Health, Klecany 25067, Czech Republic; Department of Psychology and Life Sciences, Faculty of Humanities, Charles University, Prague 18200, Czech Republic
| | - Marcos García-Juárez
- Centro de Investigación de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
| | - Raymundo Domínguez Ordóñez
- Centro de Investigación de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México; Licenciatura en Ingeniería Agronómica y Zootecnia, Complejo Regional Centro, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | | | - Rosa Angélica Lucio
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Oscar González-Flores
- Centro de Investigación de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México.
| |
Collapse
|
3
|
Charmanna SO, Srinivas A, Prashant A, Jain A, Kishor M, Shivashankar KK. PvuII-ESR1 gene polymorphism in premenstrual dysphoric disorder in South Indian women. Indian J Psychiatry 2024; 66:911-917. [PMID: 39668871 PMCID: PMC11633254 DOI: 10.4103/indianjpsychiatry.indianjpsychiatry_461_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 12/14/2024] Open
Abstract
Background Premenstrual dysphoric disorder (PMDD) is a condition that affects nearly 3-9% of the women in the reproductive age during the luteal phase of each menstrual cycle characterized by symptoms varying in severity and affecting the quality of life. Earlier research studies conducted have reported independent relationships between PvuII-ESR1-polymorphism and psychological traits in PMDD and risk for cognitive, behavioral, and affective symptoms. However, as the studies are few in number and the results are not consistent, there is a need for our study to link between the PvuII-ESR1gene and PMDD. Methodology All nonpregnant women aged between 18 and 45 years and attending the OBG or Medicine or Psychiatry OPD for a routine health checkup were recruited into the study. The cross-sectional study recruited 35 samples each in the control and PMDD groups using a validated screening PMDD Assessment Scale Questionnaire (PMDDASQ). Mann-Whitney's U test and Chi-square test were used to calculate P values for the continuous and categorical variables. Tetra-primer-amplification refractory mutation system-polymerase chain reaction was used to identify the PvuII-ESR1gene polymorphism after isolation of genomic DNA from the whole blood. Results Data-Analysis Pak tool and Med-Calc software were used for data analysis. The PvuII-ESR1 genotype distribution was in Hardy-Weinberg equilibrium in both PMDD and controls. The PMDDASQ scoring showed a significance with P ≤ 0.05. Pearson's Chi-Squared test performed for genotypes and alleles did not show any significant association with the phenotype. Conclusion PvuII-ESR1 SNP (T/C rs2234693) does not show any association with phenotype between the control and PMDD. However, PMDDAS questionnaire can be used to differentiate the women who are controls from PMDD.
Collapse
Affiliation(s)
- Shuchi Odiyanda Charmanna
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | - Anju Srinivas
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | - Akila Prashant
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | - Asmita Jain
- Department of Clinical Psychology, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | - M Kishor
- Department of Psychiatry, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | - Kusuma K. Shivashankar
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| |
Collapse
|
4
|
Proffitt MR, Smith GT. Species variation in steroid hormone-related gene expression contributes to species diversity in sexually dimorphic communication in electric fishes. Horm Behav 2024; 164:105576. [PMID: 38852479 PMCID: PMC11330740 DOI: 10.1016/j.yhbeh.2024.105576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Sexually dimorphic behaviors are often regulated by gonadal steroid hormones. Species diversity in behavioral sex differences may arise as expression of genes mediating steroid action in brain regions controlling these behaviors evolves. The electric communication signals of apteronotid knifefishes are an excellent model for comparatively studying neuroendocrine regulation of sexually dimorphic behavior. These fish produce and detect weak electric organ discharges (EODs) for electrolocation and communication. EOD frequency (EODf), controlled by the medullary pacemaker nucleus (Pn), is sexually dimorphic and regulated by androgens and estrogens in some species, but is sexually monomorphic and unaffected by hormones in other species. We quantified expression of genes for steroid receptors, metabolizing enzymes, and cofactors in the Pn of two species with sexually dimorphic EODf (Apteronotus albifrons and Apteronotus leptorhynchus) and two species with sexually monomorphic EODf ("Apteronotus" bonapartii and Parapteronotus hasemani). The "A." bonapartii Pn expressed lower levels of androgen receptor (AR) genes than the Pn of species with sexually dimorphic EODf. In contrast, the P. hasemani Pn robustly expressed AR genes, but expressed lower levels of genes for 5α-reductases, which convert androgens to more potent metabolites, and higher levels of genes for 17β-hydroxysteroid dehydrogenases that oxidize androgens and estrogens to less potent forms. These findings suggest that sexual monomorphism of EODf arose convergently via two different mechanisms. In "A." bonapartii, reduced Pn expression of ARs likely results in insensitivity of EODf to androgens, whereas in P. hasemani, gonadal steroids may be metabolically inactivated in the Pn, reducing their potential to influence EODf.
Collapse
Affiliation(s)
- Melissa R Proffitt
- Department of Biology, Indiana University, 1001 E. 3(rd) St., Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, 409 N. Park Ave., Bloomington, IN 47505, USA
| | - G Troy Smith
- Department of Biology, Indiana University, 1001 E. 3(rd) St., Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, 409 N. Park Ave., Bloomington, IN 47505, USA.
| |
Collapse
|
5
|
Acharya KD, Graham M, Raman H, Parakoyi AER, Corcoran A, Belete M, Ramaswamy B, Koul S, Sachar I, Derendorf K, Wilmer JB, Gottipati S, Tetel MJ. Estradiol-mediated protection against high-fat diet induced anxiety and obesity is associated with changes in the gut microbiota in female mice. Sci Rep 2023; 13:4776. [PMID: 36959275 PMCID: PMC10036463 DOI: 10.1038/s41598-023-31783-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/17/2023] [Indexed: 03/25/2023] Open
Abstract
Decreased estrogens during menopause are associated with increased risk of anxiety, depression, type 2 diabetes and obesity. Similarly, depleting estrogens in rodents by ovariectomy, combined with a high-fat diet (HFD), increases anxiety and adiposity. How estrogens and diet interact to affect anxiety and metabolism is poorly understood. Mounting evidence indicates that gut microbiota influence anxiety and metabolism. Here, we investigated the effects of estradiol (E) and HFD on anxiety, metabolism, and their correlation with changes in gut microbiota in female mice. Adult C57BL/6J mice were ovariectomized, implanted with E or vehicle-containing capsules and fed a standard diet or HFD. Anxiety-like behavior was assessed and neuronal activation was measured by c-fos immunoreactivity throughout the brain using iDISCO. HFD increased anxiety-like behavior, while E reduced this HFD-dependent anxiogenic effect. Interestingly, E decreased neuronal activation in brain regions involved in anxiety and metabolism. E treatment also altered gut microbes, a subset of which were associated with anxiety-like behavior. These findings provide insight into gut microbiota-based therapies for anxiety and metabolic disorders associated with declining estrogens in menopausal women.
Collapse
Affiliation(s)
- Kalpana D Acharya
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Madeline Graham
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Harshini Raman
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | | | - Alexis Corcoran
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Merzu Belete
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Bharath Ramaswamy
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Shashikant Koul
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | | | - Kevin Derendorf
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Jeremy B Wilmer
- Department of Psychology, Wellesley College, Wellesley, MA, 02481, USA
| | - Srikanth Gottipati
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Marc J Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA.
| |
Collapse
|
6
|
Hanna Mossad Samaan M, Mohamed Ahmed Sarhan T, Abd El Azim Ammar R, Hanafy Mahmoud T, Mohamed Ahmed El Shafie A. A study of menstrual cycle effects on pain perception, haemodynamic response to laryngoscopy, and postoperative outcome in gynaecological laparoscopy. EGYPTIAN JOURNAL OF ANAESTHESIA 2022. [DOI: 10.1080/11101849.2022.2147472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Mariam Hanna Mossad Samaan
- Assistant Lecturer in Anaesthesia and Surgical Intensive Care, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Tarek Mohamed Ahmed Sarhan
- Assistant Lecturer in Anaesthesia and Surgical Intensive Care, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Ramadan Abd El Azim Ammar
- Assistant Lecturer in Anaesthesia and Surgical Intensive Care, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Tamer Hanafy Mahmoud
- Professor of Obstetrics and Gynaecology, Alexandria Faculty of Medicine, Alexandria, Egypt
| | | |
Collapse
|
7
|
Zhou Y, Yan H, Liu W, Hu C, Zhou Y, Sun R, Tang Y, Zheng C, Yang J, Cui Q. A multi-tissue transcriptomic landscape of female mice in estrus and diestrus provides clues for precision medicine. Front Cell Dev Biol 2022; 10:983712. [PMID: 36589755 PMCID: PMC9800588 DOI: 10.3389/fcell.2022.983712] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Female reproductive cycle, also known as menstrual cycle or estrous cycle in primate or non-primate mammals, respectively, dominates the reproductive processes in non-pregnant state. However, in addition to reproductive tissues, reproductive cycle could also perform global regulation because the receptors of two major female hormones fluctuating throughout the cycle, estrogen and progesterone, are widely distributed. Therefore, a multi-tissue gene expression landscape is in continuous demand for better understanding the systemic changes during the reproductive cycle but remains largely undefined. Here we delineated a transcriptomic landscape covering 15 tissues of C57BL/6J female mice in two phases of estrous cycle, estrus and diestrus, by RNA-sequencing. Then, a number of genes, pathways, and transcription factors involved in the estrous cycle were revealed. We found the estrous cycle could widely regulate the neuro-functions, immuno-functions, blood coagulation and so on. And behind the transcriptomic alteration between estrus and diestrus, 13 transcription factors may play important roles. Next, bioinformatics modeling with 1,263 manually curated gene signatures of various physiological and pathophysiological states systematically characterized the beneficial/deleterious effects brought by estrus/diestrus on individual tissues. We revealed that the estrous cycle has a significant effect on cardiovascular system (aorta, heart, vein), in which the anti-hypertensive pattern in aorta induced by estrus is one of the most striking findings. Inspired by this point, we validated that two hypotensive drugs, felodipine and acebutolol, could exhibit significantly enhanced efficacy in estrus than diestrus by mouse and rat experiments. Together, this study provides a valuable data resource for investigating reproductive cycle from a transcriptomic perspective, and presents models and clues for investigating precision medicine associated with reproductive cycle.
Collapse
Affiliation(s)
- Yiran Zhou
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China,Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China,Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Han Yan
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenjun Liu
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chengqing Hu
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China,Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ruya Sun
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China,Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yida Tang
- Department of Cardiology, MOE Key Lab of Cardiovascular Sciences, Peking University Third Hospital, Beijing, China
| | - Chao Zheng
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China,*Correspondence: Chao Zheng, ; Jichun Yang, ; Qinghua Cui,
| | - Jichun Yang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China,*Correspondence: Chao Zheng, ; Jichun Yang, ; Qinghua Cui,
| | - Qinghua Cui
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China,Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China,*Correspondence: Chao Zheng, ; Jichun Yang, ; Qinghua Cui,
| |
Collapse
|
8
|
Kang S, Jo H, Kim MR. Safety Assessment of Endocrine Disruption by Menopausal Health Functional Ingredients. Healthcare (Basel) 2021; 9:healthcare9101376. [PMID: 34683056 PMCID: PMC8544397 DOI: 10.3390/healthcare9101376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 12/23/2022] Open
Abstract
During menopause, women experience various symptoms including hot flashes, mood changes, insomnia, and sweating. Hormone replacement therapy (HRT) has been used as the main treatment for menopausal symptoms; however, other options are required for women with medical contraindications or without preference for HRT. Functional health foods are easily available options for relieving menopausal symptoms. There are growing concerns regarding menopausal functional health foods because the majority of them include phytoestrogens which have the effect of endocrine disruption. Phytoestrogens may cause not only hormonal imbalance or disruption of the normal biological function of the organ systems, but also uterine cancer or breast cancer if absorbed and accumulated in the body for a long period of time, depending on the estrogen receptor binding capacity. Therefore, we aimed to determine the effects and safety of menopausal functional health ingredients and medicines on the human body as endocrine disruptors under review in the literature and the OECD guidelines.
Collapse
Affiliation(s)
- Soyeon Kang
- St. Vincent’s Hospital, Division of Gynecologic Endocrinology, Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Hagyeong Jo
- Seoul St. Mary’s Hospital, Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Mee-Ran Kim
- Seoul St. Mary’s Hospital, Division of Gynecologic Endocrinology, Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-6170
| |
Collapse
|
9
|
Acharya KD, Noh HL, Graham ME, Suk S, Friedline RH, Gomez CC, Parakoyi AER, Chen J, Kim JK, Tetel MJ. Distinct Changes in Gut Microbiota Are Associated with Estradiol-Mediated Protection from Diet-Induced Obesity in Female Mice. Metabolites 2021; 11:metabo11080499. [PMID: 34436440 PMCID: PMC8398128 DOI: 10.3390/metabo11080499] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 01/14/2023] Open
Abstract
A decrease in ovarian estrogens in postmenopausal women increases the risk of weight gain, cardiovascular disease, type 2 diabetes, and chronic inflammation. While it is known that gut microbiota regulates energy homeostasis, it is unclear if gut microbiota is associated with estradiol regulation of metabolism. In this study, we tested if estradiol-mediated protection from high-fat diet (HFD)-induced obesity and metabolic changes are associated with longitudinal alterations in gut microbiota in female mice. Ovariectomized adult mice with vehicle or estradiol (E2) implants were fed chow for two weeks and HFD for four weeks. As reported previously, E2 increased energy expenditure, physical activity, insulin sensitivity, and whole-body glucose turnover. Interestingly, E2 decreased the tight junction protein occludin, suggesting E2 affects gut epithelial integrity. Moreover, E2 increased Akkermansia and decreased Erysipleotrichaceae and Streptococcaceae. Furthermore, Coprobacillus and Lactococcus were positively correlated, while Akkermansia was negatively correlated, with body weight and fat mass. These results suggest that changes in gut epithelial barrier and specific gut microbiota contribute to E2-mediated protection against diet-induced obesity and metabolic dysregulation. These findings provide support for the gut microbiota as a therapeutic target for treating estrogen-dependent metabolic disorders in women.
Collapse
Affiliation(s)
- Kalpana D. Acharya
- Neuroscience Department, Wellesley College, Wellesley, MA 02481, USA; (K.D.A.); (M.E.G.); (C.C.G.); (A.E.R.P.)
| | - Hye L. Noh
- Program in Molecular Medicine, Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; (H.L.N.); (S.S.); (R.H.F.); (J.K.K.)
| | - Madeline E. Graham
- Neuroscience Department, Wellesley College, Wellesley, MA 02481, USA; (K.D.A.); (M.E.G.); (C.C.G.); (A.E.R.P.)
| | - Sujin Suk
- Program in Molecular Medicine, Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; (H.L.N.); (S.S.); (R.H.F.); (J.K.K.)
| | - Randall H. Friedline
- Program in Molecular Medicine, Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; (H.L.N.); (S.S.); (R.H.F.); (J.K.K.)
| | - Cesiah C. Gomez
- Neuroscience Department, Wellesley College, Wellesley, MA 02481, USA; (K.D.A.); (M.E.G.); (C.C.G.); (A.E.R.P.)
| | - Abigail E. R. Parakoyi
- Neuroscience Department, Wellesley College, Wellesley, MA 02481, USA; (K.D.A.); (M.E.G.); (C.C.G.); (A.E.R.P.)
| | - Jun Chen
- Department of Health Sciences Research & Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Jason K. Kim
- Program in Molecular Medicine, Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; (H.L.N.); (S.S.); (R.H.F.); (J.K.K.)
| | - Marc J. Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA 02481, USA; (K.D.A.); (M.E.G.); (C.C.G.); (A.E.R.P.)
- Correspondence:
| |
Collapse
|
10
|
Chiang VSC, Park JH. Glutamate in Male and Female Sexual Behavior: Receptors, Transporters, and Steroid Independence. Front Behav Neurosci 2020; 14:589882. [PMID: 33328921 PMCID: PMC7732465 DOI: 10.3389/fnbeh.2020.589882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/20/2020] [Indexed: 01/12/2023] Open
Abstract
The survival of animal species predicates on the success of sexual reproduction. Neurotransmitters play an integral role in the expression of these sexual behaviors in the brain. Here, we review the role of glutamate in sexual behavior in rodents and non-rodent species for both males and females. These encompass the release of glutamate and correlations with glutamate receptor expression during sexual behavior. We then present the effects of glutamate on sexual behavior, as well as the effects of antagonists and agonists on different glutamate transporters and receptors. Following that, we discuss the potential role of glutamate on steroid-independent sexual behavior. Finally, we demonstrate the interaction of glutamate with other neurotransmitters to impact sexual behavior. These sexual behavior studies are crucial in the development of novel treatments of sexual dysfunction and in furthering our understanding of the complexity of sexual diversity. In the past decade, we have witnessed the burgeoning of novel techniques to study and manipulate neuron activity, to decode molecular events at the single-cell level, and to analyze behavioral data. They pose exciting avenues to gain further insight into future sexual behavior research. Taken together, this work conveys the essential role of glutamate in sexual behavior.
Collapse
Affiliation(s)
- Vic Shao-Chih Chiang
- Developmental and Brain Sciences, Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
| | - Jin Ho Park
- Developmental and Brain Sciences, Department of Psychology, University of Massachusetts Boston, Boston, MA, United States
| |
Collapse
|
11
|
Acharya KD, Gao X, Bless EP, Chen J, Tetel MJ. Estradiol and high fat diet associate with changes in gut microbiota in female ob/ob mice. Sci Rep 2019; 9:20192. [PMID: 31882890 PMCID: PMC6934844 DOI: 10.1038/s41598-019-56723-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022] Open
Abstract
Estrogens protect against diet-induced obesity in women and female rodents. For example, a lack of estrogens in postmenopausal women is associated with an increased risk of weight gain, cardiovascular diseases, low-grade inflammation, and cancer. Estrogens act with leptin to regulate energy homeostasis in females. Leptin-deficient mice (ob/ob) exhibit morbid obesity and insulin resistance. The gut microbiome is also critical in regulating metabolism. The present study investigates whether estrogens and leptin modulate gut microbiota in ovariectomized ob/ob (obese) or heterozygote (lean) mice fed high-fat diet (HFD) that received either 17β-Estradiol (E2) or vehicle implants. E2 attenuated weight gain in both genotypes. Moreover, both obesity (ob/ob mice) and E2 were associated with reduced gut microbial diversity. ob/ob mice exhibited lower species richness than control mice, while E2-treated mice had reduced evenness compared with vehicle mice. Regarding taxa, E2 was associated with an increased abundance of the S24-7 family, while leptin was associated with increases in Coriobacteriaceae, Clostridium and Lactobacillus. Some taxa were affected by both E2 and leptin, suggesting these hormones alter gut microbiota of HFD-fed female mice. Understanding the role of E2 and leptin in regulating gut microbiota will provide important insights into hormone-dependent metabolic disorders in women.
Collapse
Affiliation(s)
- Kalpana D Acharya
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA.
| | - Xing Gao
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Elizabeth P Bless
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Jun Chen
- Department of Health Sciences Research & Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Marc J Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| |
Collapse
|
12
|
Neonatal treatment with clomipramine modifies the expression of estrogen receptors in brain areas of male adult rats. Brain Res 2019; 1724:146443. [DOI: 10.1016/j.brainres.2019.146443] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/04/2019] [Accepted: 09/08/2019] [Indexed: 01/03/2023]
|
13
|
Jaric I, Rocks D, Greally JM, Suzuki M, Kundakovic M. Chromatin organization in the female mouse brain fluctuates across the oestrous cycle. Nat Commun 2019; 10:2851. [PMID: 31253786 PMCID: PMC6598989 DOI: 10.1038/s41467-019-10704-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 05/23/2019] [Indexed: 12/12/2022] Open
Abstract
Male and female brains differ significantly in both health and disease, and yet the female brain has been understudied. Sex-hormone fluctuations make the female brain particularly dynamic and are likely to confer female-specific risks for neuropsychiatric disorders. The molecular mechanisms underlying the dynamic nature of the female brain structure and function are unknown. Here we show that neuronal chromatin organization in the female ventral hippocampus of mouse fluctuates with the oestrous cycle. We find chromatin organizational changes associated with the transcriptional activity of genes important for neuronal function and behaviour. We link these chromatin dynamics to variation in anxiety-related behaviour and brain structure. Our findings implicate an immediate-early gene product, Egr1, as part of the mechanism mediating oestrous cycle-dependent chromatin and transcriptional changes. This study reveals extreme, sex-specific dynamism of the neuronal epigenome, and establishes a foundation for the development of sex-specific treatments for disorders such as anxiety and depression.
Collapse
Affiliation(s)
- Ivana Jaric
- Department of Biological Sciences, Fordham University, 441 E. Fordham Road, Bronx, NY, 10458, USA
| | - Devin Rocks
- Department of Biological Sciences, Fordham University, 441 E. Fordham Road, Bronx, NY, 10458, USA
| | - John M Greally
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Masako Suzuki
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Marija Kundakovic
- Department of Biological Sciences, Fordham University, 441 E. Fordham Road, Bronx, NY, 10458, USA.
| |
Collapse
|
14
|
Emerging Roles of Estrogen-Related Receptors in the Brain: Potential Interactions with Estrogen Signaling. Int J Mol Sci 2018; 19:ijms19041091. [PMID: 29621182 PMCID: PMC5979530 DOI: 10.3390/ijms19041091] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/21/2018] [Accepted: 03/30/2018] [Indexed: 01/22/2023] Open
Abstract
In addition to their well-known role in the female reproductive system, estrogens can act in the brain to regulate a wide range of behaviors and physiological functions in both sexes. Over the past few decades, genetically modified animal models have greatly increased our knowledge about the roles of estrogen receptor (ER) signaling in the brain in behavioral and physiological regulations. However, less attention has been paid to the estrogen-related receptors (ERRs), the members of orphan nuclear receptors whose sequences are homologous to ERs but lack estrogen-binding ability. While endogenous ligands of ERRs remain to be determined, they seemingly share transcriptional targets with ERs and their expression can be directly regulated by ERs through the estrogen-response element embedded within the regulatory region of the genes encoding ERRs. Despite the broad expression of ERRs in the brain, we have just begun to understand the fundamental roles they play at molecular, cellular, and circuit levels. Here, we review recent research advancement in understanding the roles of ERs and ERRs in the brain, with particular emphasis on ERRs, and discuss possible cross-talk between ERs and ERRs in behavioral and physiological regulations.
Collapse
|
15
|
Kundakovic M. Sex-Specific Epigenetics: Implications for Environmental Studies of Brain and Behavior. Curr Environ Health Rep 2018; 4:385-391. [PMID: 28986864 DOI: 10.1007/s40572-017-0172-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW This review discusses the current state of knowledge on sex differences in the epigenetic regulation in the brain and highlights its relevance for the environmental studies of brain and behavior. RECENT FINDINGS Recent evidence shows that epigenetic mechanisms are involved in the control of brain sexual differentiation and in memory-enhancing effects of estradiol in females. In addition, several studies have implicated epigenetic dysregulation as an underlying mechanism for sex-specific neurobehavioral effects of environmental exposures. The area of sex-specific neurepigenetics has a great potential to improve our understanding of brain function in health and disease. Future neuropigenetic studies will require the inclusion of males and females and would ideally account for the fluctuating hormonal status in females which is likely to affect the epigenome. The implementation of cutting-edge methods that include epigenomic characterization of specific cell types using latest next-generation sequencing approaches will further advance the area.
Collapse
Affiliation(s)
- Marija Kundakovic
- Department of Biological Sciences, Fordham University, 441 E. Fordham Road, Larkin Hall, Room 160, Bronx, NY, 10458, USA.
| |
Collapse
|
16
|
Tetel MJ, de Vries GJ, Melcangi RC, Panzica G, O'Mahony SM. Steroids, stress and the gut microbiome-brain axis. J Neuroendocrinol 2018; 30:10.1111/jne.12548. [PMID: 29024170 PMCID: PMC6314837 DOI: 10.1111/jne.12548] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/07/2017] [Accepted: 10/09/2017] [Indexed: 12/14/2022]
Abstract
It is becoming well established that the gut microbiome has a profound impact on human health and disease. In this review, we explore how steroids can influence the gut microbiota and, in turn, how the gut microbiota can influence hormone levels. Within the context of the gut microbiome-brain axis, we discuss how perturbations in the gut microbiota can alter the stress axis and behaviour. In addition, human studies on the possible role of gut microbiota in depression and anxiety are examined. Finally, we present some of the challenges and important questions that need to be addressed by future research in this exciting new area at the intersection of steroids, stress, gut-brain axis and human health.
Collapse
Affiliation(s)
- M J Tetel
- Neuroscience Program, Wellesley College, Wellesley, MA, USA
| | - G J de Vries
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - R C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - G Panzica
- Dipartimento di Neuroscienze "Rita Levi Montalcini", Neuroscience Institute Cavalieri Ottolenghi (NICO), Università degli Studi di Torino, Orbassano, Italy
| | - S M O'Mahony
- Department of Anatomy and Neuroscience, APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
17
|
Ohtani N, Suda K, Tsuji E, Tanemura K, Yokota H, Inoue H, Iwano H. Late pregnancy is vulnerable period for exposure to BPA. J Vet Med Sci 2018; 80:536-543. [PMID: 29367495 PMCID: PMC5880839 DOI: 10.1292/jvms.17-0460] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bisphenol A (BPA) is among the better-known endocrine disruptors. BPA is used in various food-contacting materials and is easily eluted into food; as a result, we are exposed to BPA on a daily basis. In adults, BPA is
metabolized and eliminated rapidly from the body. However, numerous reports suggest that fetuses and young children are susceptible to BPA. One of the concerning adverse effects of BPA is disruption of behavior,
especially anxiety-like behavior. In order to study the mechanism of influences on offspring, it is important to clarify the most vulnerable gestation period. We hypothesized that offspring in late pregnancy would be
more susceptible to BPA, because late pregnancy is a critical time for functional brain development. In this study, C57BL/6 mouse fetuses were exposed prenatally by oral dosing of pregnant dams, once daily from
gestational day 5.5 to 12.5 (early pregnancy) or 11.5 to 18.5 (late pregnancy), with BPA (0 or 10 mg/kg body weight). Following birth and weaning, the resulting pups were tested using an elevated plus maze at postnatal
week 10. The behavior of the offspring was altered by prenatal BPA exposure during late pregnancy but not during early pregnancy. These results indicated that offspring are more vulnerable to exposure to BPA in late
pregnancy.
Collapse
Affiliation(s)
- Naoko Ohtani
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Koshi Suda
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Erika Tsuji
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Kentaro Tanemura
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 981-8555, Japan
| | - Hiroshi Yokota
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Hiroki Inoue
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Hidetomo Iwano
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| |
Collapse
|
18
|
Arambula SE, Jima D, Patisaul HB. Prenatal bisphenol A (BPA) exposure alters the transcriptome of the neonate rat amygdala in a sex-specific manner: a CLARITY-BPA consortium study. Neurotoxicology 2017; 65:207-220. [PMID: 29097150 DOI: 10.1016/j.neuro.2017.10.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/11/2022]
Abstract
Bisphenol A (BPA) is a widely recognized endocrine disruptor prevalent in many household items. Because experimental and epidemiological data suggest links between prenatal BPA exposure and altered affective behaviors in children, even at levels below the current US FDA No Observed Adverse Effect Level (NOAEL) of 5mg/kg body weight (bw)/day, there is concern that early life exposure may alter neurodevelopment. The current study was conducted as part of the CLARITY-BPA (Consortium Linking Academic and Regulatory Insights on BPA Toxicity) program and examined the full amygdalar transcriptome on postnatal day (PND) 1, with the hypothesis that prenatal BPA exposure would alter the expression of genes and pathways fundamental to sex-specific affective behaviors. NCTR Sprague-Dawley dams were gavaged from gestational day 6 until parturition with BPA (2.5, 25, 250, 2500, or 25000μg/kg bw/day), a reference estrogen (0.05 or 0.5μg ethinyl estradiol (EE2)/kg bw/day), or vehicle. PND 1 amygdalae were microdissected and gene expression was assessed with qRT-PCR (all exposure groups) and RNAseq (vehicle, 25 and 250μg BPA, and 0.5μg EE2 groups only). Our results demonstrate that that prenatal BPA exposure can disrupt the transcriptome of the neonate amygdala, at doses below the FDA NOAEL, in a sex-specific manner and indicate that the female amygdala may be more sensitive to BPA exposure during fetal development. We also provide additional evidence that developmental BPA exposure can interfere with estrogen, oxytocin, and vasopressin signaling pathways in the developing brain and alter signaling pathways critical for synaptic organization and transmission.
Collapse
Affiliation(s)
- Sheryl E Arambula
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; WM Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA
| | - Dereje Jima
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; WM Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
19
|
Frago LM, Canelles S, Freire-Regatillo A, Argente-Arizón P, Barrios V, Argente J, Garcia-Segura LM, Chowen JA. Estradiol Uses Different Mechanisms in Astrocytes from the Hippocampus of Male and Female Rats to Protect against Damage Induced by Palmitic Acid. Front Mol Neurosci 2017; 10:330. [PMID: 29114202 PMCID: PMC5660686 DOI: 10.3389/fnmol.2017.00330] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/29/2017] [Indexed: 01/22/2023] Open
Abstract
An excess of saturated fatty acids can be toxic for tissues, including the brain, and this has been associated with the progression of neurodegenerative diseases. Since palmitic acid (PA) is a free fatty acid that is abundant in the diet and circulation and can be harmful, we have investigated the effects of this fatty acid on lipotoxicity in hippocampal astrocytes and the mechanism involved. Moreover, as males and females have different susceptibilities to some neurodegenerative diseases, we accessed the responses of astrocytes from both sexes, as well as the possible involvement of estrogens in the protection against fatty acid toxicity. PA increased endoplasmic reticulum stress leading to cell death in astrocytes from both males and females. Estradiol (E2) increased the levels of protective factors, such as Hsp70 and the anti-inflammatory cytokine interleukin-10, in astrocytes from both sexes. In male astrocytes, E2 decreased pJNK, TNFα, and caspase-3 activation. In contrast, in female astrocytes E2 did not affect the activation of JNK or TNFα levels, but decreased apoptotic cell death. Hence, although E2 exerted protective effects against the detrimental effects of PA, the mechanisms involved appear to be different between male and female astrocytes. This sexually dimorphic difference in the protective mechanisms induced by E2 could be involved in the different susceptibilities of males and females to some neurodegenerative processes.
Collapse
Affiliation(s)
- Laura M Frago
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Canelles
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Argente-Arizón
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Barrios
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain.,IMDEA Food Institute, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBER de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A Chowen
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
20
|
Estradiol increases choice of cocaine over food in male rats. Physiol Behav 2017; 203:18-24. [PMID: 29056351 DOI: 10.1016/j.physbeh.2017.10.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 12/21/2022]
Abstract
Estradiol modulates the rewarding and reinforcing properties of cocaine in females, including an increase in selection of cocaine over alternative reinforcers. However, the effects of estradiol on male cocaine self-administration behavior are less studied despite equivalent levels of estradiol in the brains of adult males and females, estradiol effects on motivated behaviors in males that share underlying neural substrates with cocaine reinforcement as well as expression of estrogen receptors in the male brain. Therefore, we sought to characterize the effects of estradiol in males on choice between concurrently-available cocaine and food reinforcement as well as responding for cocaine or food in isolation. Male castrated rats (n=46) were treated daily with estradiol benzoate (EB) (5μg/0.1, S.C.) or vehicle (peanut oil) throughout operant acquisition of cocaine (1mg/kg, IV; FI20 sec) and food (3×45mg; FI20 sec) responding, choice during concurrent access and cocaine and food reinforcement under progressive ratio (PR) schedules. EB increased cocaine choice, both in terms of percent of trials on which cocaine was selected and the proportion of rats exhibiting a cocaine preference as well as increased cocaine, but not food, intake under PR. Additionally, within the EB treated group, cocaine-preferring rats exhibited enhanced acquisition of cocaine, but not food, reinforcement whereas no acquisition differences were observed across preferences in the vehicle treated group. These findings demonstrate that estradiol increases cocaine choice in males similarly to what is observed in females.
Collapse
|
21
|
Maney DL. Polymorphisms in sex steroid receptors: From gene sequence to behavior. Front Neuroendocrinol 2017; 47:47-65. [PMID: 28705582 PMCID: PMC6312198 DOI: 10.1016/j.yfrne.2017.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/05/2017] [Accepted: 07/08/2017] [Indexed: 01/09/2023]
Abstract
Sex steroid receptors have received much interest as potential mediators of human behaviors and mental disorders. Candidate gene association studies have identified about 50 genetic variants of androgen and estrogen receptors that correlate with human behavioral phenotypes. Because most of these polymorphisms lie outside coding regions, discerning their effect on receptor function is not straightforward. Thus, although discoveries of associations improve our ability to predict risk, they have not greatly advanced our understanding of underlying mechanisms. This article is intended to serve as a starting point for psychologists and other behavioral biologists to consider potential mechanisms. Here, I review associations between polymorphisms in sex steroid receptors and human behavioral phenotypes. I then consider ways in which genetic variation can affect processes such as mRNA transcription, splicing, and stability. Finally, I suggest ways that hypotheses about mechanism can be tested, for example using in vitro assays and/or animal models.
Collapse
Affiliation(s)
- Donna L Maney
- Department of Psychology, 36 Eagle Row, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
22
|
The Epigenetic Link between Prenatal Adverse Environments and Neurodevelopmental Disorders. Genes (Basel) 2017; 8:genes8030104. [PMID: 28335457 PMCID: PMC5368708 DOI: 10.3390/genes8030104] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/08/2017] [Accepted: 03/12/2017] [Indexed: 12/12/2022] Open
Abstract
Prenatal adverse environments, such as maternal stress, toxicological exposures, and viral infections, can disrupt normal brain development and contribute to neurodevelopmental disorders, including schizophrenia, depression, and autism. Increasing evidence shows that these short- and long-term effects of prenatal exposures on brain structure and function are mediated by epigenetic mechanisms. Animal studies demonstrate that prenatal exposure to stress, toxins, viral mimetics, and drugs induces lasting epigenetic changes in the brain, including genes encoding glucocorticoid receptor (Nr3c1) and brain-derived neurotrophic factor (Bdnf). These epigenetic changes have been linked to changes in brain gene expression, stress reactivity, and behavior, and often times, these effects are shown to be dependent on the gestational window of exposure, sex, and exposure level. Although evidence from human studies is more limited, gestational exposure to environmental risks in humans is associated with epigenetic changes in peripheral tissues, and future studies are required to understand whether we can use peripheral biomarkers to predict neurobehavioral outcomes. An extensive research effort combining well-designed human and animal studies, with comprehensive epigenomic analyses of peripheral and brain tissues over time, will be necessary to improve our understanding of the epigenetic basis of neurodevelopmental disorders.
Collapse
|
23
|
Domínguez-Ordóñez R, García-Juárez M, Lima-Hernández FJ, Gómora-Arrati P, Blaustein JD, Etgen AM, González-Flores O. Estrogen receptor α and β are involved in the activation of lordosis behavior in estradiol-primed rats. Horm Behav 2016; 86:1-7. [PMID: 27594441 DOI: 10.1016/j.yhbeh.2016.08.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 08/28/2016] [Accepted: 08/31/2016] [Indexed: 11/17/2022]
Abstract
The present study was designed to assess the participation of estrogen receptors alpha (ERα) and beta (ERβ) in the short-term facilitation of lordosis behavior in ovariectomized (ovx), estradiol (E2) primed rats. In experiment 1, dose response curves for PPT and DPN (ERα and ERβ agonists, respectively) facilitation of lordosis behavior (lordosis quotient and lordosis score) were established by infusing these agonists into the right lateral ventricle (icv) in female rats injected 40h previously with 5μg of E2 benzoate. PPT doses of 0.08 and 0.4ng produced high lordosis quotients starting at 30min and continuing at 120 and 240min post-injection. DPN induced high levels of lordosis behavior at all times tested. However, the intensity of lordosis induced by both agonists was weak. In experiment 2, we tested the involvement of each ER in facilitation of lordosis by icv infusion of MPP (ERα-selective antagonist) or PHTPP (ERβ-selective antagonist) prior to infusion of 2ng of free E2. Icv infusion of either MPP or PHTPP 30min before free E2 significantly depressed E2 facilitation of lordosis. The results suggest that both forms of ER are involved in the short-latency facilitation of lordosis behavior in E2-primed rats.
Collapse
Affiliation(s)
- Raymundo Domínguez-Ordóñez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México; Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Marcos García-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México
| | - Francisco J Lima-Hernández
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México; Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tlaxcala, México
| | - Porfirio Gómora-Arrati
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México
| | - Jeffrey D Blaustein
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Anne M Etgen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México.
| |
Collapse
|
24
|
Zhang R, Hu Y, Wang H, Yan P, Zhou Y, Wu R, Wu X. Molecular cloning, characterization, tissue distribution and mRNA expression changes during the hibernation and reproductive periods of estrogen receptor alpha (ESR1) in Chinese alligator, Alligator sinensis. Comp Biochem Physiol B Biochem Mol Biol 2016; 200:28-35. [PMID: 27212643 DOI: 10.1016/j.cbpb.2016.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 02/07/2023]
Abstract
Chinese alligator, Alligator sinensis, is a critically endangered reptile species unique to China. Little is known about the mechanism of growth- and reproduction-related hormones gene expression in Chinese alligator. Estrogens play important roles in regulating multiple reproduction- and non-reproduction-related functions by binding to their corresponding receptors. Here, the full-length cDNA of estrogen receptor alpha (ERα/ESR1) was cloned and sequenced from Chinese alligator for the first time, which comprises 1764bp nucleotides and encodes a predicted protein of 587 amino acids. Phylogenetic analysis of ESR1 showed that crocodilians and turtles were the sister-group of birds. The results of real-time quantitative PCR indicated that the ESR1 mRNA was widely expressed in the brain and peripheral tissues. In the brain and pituitary gland, ESR1 was most highly transcribed in the cerebellum. But in other peripheral tissues, ESR1 mRNA expression level was the highest in the ovary. Compared with hibernation period, ESR1 mRNA expression levels were increased significantly in the reproductive period (P<0.05) in cerebellum, pituitary gland, liver, spleen, lung, kidney and ovary, while no significant change in other examined tissues (P>0.05). The ESR1 mRNA expression levels changes during the two periods of different tissues suggested that ESR1 might play an important role in mediation of estrogenic multiple reproductive effects in Chinese alligator. Furthermore, it was the first time to quantify ESR1 mRNA level in the brain of crocodilians, and the distribution and expression of ESR1 mRNA in the midbrain, cerebellum and medulla oblongata was also reported for the first time in reptiles.
Collapse
Affiliation(s)
- Ruidong Zhang
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China
| | - Yuehong Hu
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China
| | - Huan Wang
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China
| | - Peng Yan
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China
| | - Yongkang Zhou
- Alligator Research Center of Anhui Province, Xuanzhou 242000, People's Republic of China
| | - Rong Wu
- Alligator Research Center of Anhui Province, Xuanzhou 242000, People's Republic of China
| | - Xiaobing Wu
- Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, People's Republic of China.
| |
Collapse
|
25
|
Lubec G, Korz V. Concerted Gene Expression of Hippocampal Steroid Receptors during Spatial Learning in Male Wistar Rats: A Correlation Analysis. Front Behav Neurosci 2016; 10:94. [PMID: 27242463 PMCID: PMC4868845 DOI: 10.3389/fnbeh.2016.00094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/29/2016] [Indexed: 11/29/2022] Open
Abstract
Adrenal and gonadal steroid receptor activities are significantly involved and interact in the regulation of learning, memory and stress. Thus, a coordinated expression of steroid receptor genes during a learning task can be expected. Although coexpression of steroid receptors in response to behavioral tasks has been reported the correlative connection is unclear. According to the inverted U-shape model of the impact of stress upon learning and memory we hypothesized that glucocorticoid (GR) receptor expression should be correlated to corticosterone levels in a linear or higher order manner. Other cognition modulating steroid receptors like estrogen receptors (ER) should be correlated to GR receptors in a quadratic manner, which describes a parabola and thus a U-shaped connection. Therefore, we performed a correlational meta-analyis of data of a previous study (Meyer and Korz, 2013a) of steroid receptor gene expressions during spatial learning, which provides a sufficient data basis in order to perform such correlational connections. In that study male rats of different ages were trained in a spatial holeboard or remained untrained and the hippocampal gene expression of different steroid receptors as well as serum corticosterone levels were measured. Expressions of mineralocorticoid (MR) and GR receptors were positively and linearly correlated with blood serum corticosterone levels in spatially trained but not in untrained animals. Training induced a cubic (best fit) relationship between mRNA levels of estrogen receptor α (ERα) and androgen receptor (AR) with MR mRNA. GR gene expression was linearly correlated with MR expression under both conditions. ERα m RNA levels were negatively and linearily and MR and GR gene expressions were cubicely correlated with reference memory errors (RME). Due to only three age classes correlations with age could not be performed. The findings support the U-shape theory of steroid receptor interaction, however the cubic fit suggest a more complex situation, which mechanisms may be revealed in further studies.
Collapse
Affiliation(s)
- Gert Lubec
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna Vienna, Austria
| | - Volker Korz
- Department of Pediatrics, Medical University of Vienna Vienna, Austria
| |
Collapse
|
26
|
Abstract
The estrogen receptors, ERα, ERβ, and GPER, mediate the effects of estrogenic compounds on their target tissues. Estrogen receptors are located in the tissues of the female reproductive tract and breast as one would expect, but also in tissues as diverse as bone, brain, liver, colon, skin, and salivary gland. The purpose of this discussion of the estrogen receptors is to provide a brief overview of the estrogen receptors and estrogen action from perspectives such as the historical, physiological, pharmacological, pathological, structural, and ligand perspectives.
Collapse
Affiliation(s)
- Kathleen M Eyster
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD, 57069, USA.
| |
Collapse
|
27
|
Estradiol Preferentially Induces Progestin Receptor-A (PR-A) Over PR-B in Cells Expressing Nuclear Receptor Coactivators in the Female Mouse Hypothalamus. eNeuro 2015; 2:eN-NWR-0012-15. [PMID: 26465008 PMCID: PMC4596027 DOI: 10.1523/eneuro.0012-15.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 07/24/2015] [Accepted: 07/25/2015] [Indexed: 11/29/2022] Open
Abstract
Estrogens act in brain to profoundly influence neurogenesis, sexual differentiation, neuroprotection, cognition, energy homeostasis, and female reproductive behavior and physiology through a variety of mechanisms, including the induction of progestin receptors (PRs). PRs are expressed as two isoforms, PR-A and PR-B, that have distinct functions in physiology and behavior. Because these PR isoforms cannot be distinguished using cellular resolution techniques, the present study used isoform-specific null mutant mice that lack PR-A or PR-B for the first time to investigate whether 17β-estradiol benzoate (EB) regulates the differential expression of the PR isoforms in the ventromedial nucleus of the hypothalamus (VMN), arcuate nucleus, and medial preoptic area, brain regions that are rich in EB-induced PRs. Interestingly, EB induced more PR-A than PR-B in all three brain regions, suggesting that PR-A is the predominant isoform in these regions. Given that steroid receptor coactivator (SRC)-1 and SRC-2 are important in estrogen receptor (ER)-dependent transcription in brain, including PR induction, we tested whether the expression of these coactivators was correlated with PR isoform expression. The majority of EB-induced PR cells expressed both SRC-1 and SRC-2 in the three brain regions of all genotypes. Interestingly, the intensity of PR-A immunoreactivity correlated with SRC-2 expression in the VMN, providing a potential mechanism for selective ER-mediated transactivation of PR-A over PR-B in a brain region-specific manner. In summary, these novel findings indicate that estrogens differentially regulate PR-A and PR-B expression in the female hypothalamus, and provide a mechanism by which steroid action in brain can selectively modulate behavior and physiology.
Collapse
|
28
|
Bless EP, Reddy T, Acharya KD, Beltz BS, Tetel MJ. Oestradiol and diet modulate energy homeostasis and hypothalamic neurogenesis in the adult female mouse. J Neuroendocrinol 2014; 26:805-16. [PMID: 25182179 PMCID: PMC4476296 DOI: 10.1111/jne.12206] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/27/2014] [Accepted: 08/27/2014] [Indexed: 12/19/2022]
Abstract
Leptin and oestradiol have overlapping functions in energy homeostasis and fertility, and receptors for these hormones are localised in the same hypothalamic regions. Although, historically, it was assumed that mammalian adult neurogenesis was confined to the olfactory bulbs and the hippocampus, recent research has found new neurones in the male rodent hypothalamus. Furthermore, some of these new neurones are leptin-sensitive and affected by diet. In the present study, we tested the hypothesis that diet and hormonal status modulate hypothalamic neurogenesis in the adult female mouse. Adult mice were ovariectomised and implanted with capsules containing oestradiol (E2 ) or oil. Within each group, mice were fed a high-fat diet (HFD) or maintained on standard chow (STND). All animals were administered i.c.v. 5-bromo-2'-deoxyuridine (BrdU) for 9 days and sacrificed 34 days later after an injection of leptin to induce phosphorylation of signal transducer of activation and transcription 3 (pSTAT3). Brain tissue was immunohistochemically labelled for BrdU (newly born cells), Hu (neuronal marker) and pSTAT3 (leptin sensitive). Although mice on a HFD became obese, oestradiol protected against obesity. There was a strong interaction between diet and hormone on new cells (BrdU+) in the arcuate, ventromedial hypothalamus and dorsomedial hypothalamus. HFD increased the number of new cells, whereas E2 inhibited this effect. Conversely, E2 increased the number of new cells in mice on a STND diet in all hypothalamic regions studied. Although the total number of new leptin-sensitive neurones (BrdU-Hu-pSTAT3) found in the hypothalamus was low, HFD increased these new cells in the arcuate, whereas E2 attenuated this induction. These results suggest that adult neurogenesis in the hypothalamic neurogenic niche is modulated by diet and hormonal status and is related to energy homeostasis in female mice.
Collapse
Affiliation(s)
- E P Bless
- Neuroscience Program, Wellesley College, Wellesley, MA, USA
| | | | | | | | | |
Collapse
|
29
|
Matsuda KI. Epigenetic changes in the estrogen receptor α gene promoter: implications in sociosexual behaviors. Front Neurosci 2014; 8:344. [PMID: 25389384 PMCID: PMC4211403 DOI: 10.3389/fnins.2014.00344] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 10/09/2014] [Indexed: 11/23/2022] Open
Abstract
Estrogen action through estrogen receptor α (ERα) is involved in the control of sexual and social behaviors in adult mammals. Alteration of ERα gene activity mediated by epigenetic mechanisms, such as histone modifications and DNA methylation, in particular brain areas appears to be crucial for determining the extents of these behaviors between the sexes and among individuals within the same sex. This review provides a summary of the epigenetic changes in the ERα gene promoter that correlate with sociosexual behaviors.
Collapse
Affiliation(s)
- Ken Ichi Matsuda
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kyoto, Japan
| |
Collapse
|
30
|
Meyer K, Korz V. Estrogen receptor α functions in the regulation of motivation and spatial cognition in young male rats. PLoS One 2013; 8:e79303. [PMID: 24236119 PMCID: PMC3827345 DOI: 10.1371/journal.pone.0079303] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 09/22/2013] [Indexed: 11/24/2022] Open
Abstract
Estrogenic functions in regulating behavioral states such as motivation, mood, anxiety, and cognition are relatively well documented in female humans and animals. In males, however, although the entire enzymatic machinery for producing estradiol and the corresponding receptors are present, estrogenic functions have been largely neglected. Therefore, and as a follow-up study to previous research, we sub-chronically applied a specific estrogen receptor α (ERα) antagonist in young male rats before and during a spatial learning task (holeboard). The male rats showed a dose-dependent increase in motivational, but not cognitive, behavior. The expression of hippocampal steroid receptor genes, such as glucocorticoid (GR), mineralocorticoid (MR), androgen (AR), and the estrogen receptor ERα but not ERβ was dose-dependently reduced. The expression of the aromatase but not the brain-derived neurotrophic factor (BDNF) encoding gene was also suppressed. Reduced gene expression and increased behavioral performance converged at an antagonist concentration of 7.4 µmol. The hippocampal and blood serum hormone levels (corticosterone, testosterone, and 17β-estradiol) did not differ between the experimental groups and controls. We conclude that steroid receptors (and BDNF) act in a concerted, network-like manner to affect behavior and mutual gene expression. Therefore, the isolated view on single receptor types is probably insufficient to explain steroid effects on behavior. The steroid network may keep motivation in homeostasis by supporting and constraining the behavioral expression of motivation.
Collapse
Affiliation(s)
- Katrin Meyer
- Leibniz Institute for Neurobiology, Magdeburg, Germany
- Institute for Biology, Otto-von-Guericke University, Magdeburg, Germany
| | - Volker Korz
- Institute for Biology, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Neuroscience, Magdeburg, Germany
- * E-mail:
| |
Collapse
|
31
|
Tetel MJ, Acharya KD. Nuclear receptor coactivators: regulators of steroid action in brain and behaviour. J Neuroendocrinol 2013; 25:1209-18. [PMID: 23795583 PMCID: PMC3830605 DOI: 10.1111/jne.12065] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/03/2013] [Accepted: 06/18/2013] [Indexed: 11/29/2022]
Abstract
Steroid hormones act in specific regions of the brain to alter behaviour and physiology. Although it has been well established that the bioavailability of the steroid and the expression of its receptor is critical for understanding steroid action in the brain, the importance of nuclear receptor coactivators in the brain is becoming more apparent. The present review focuses on the function of the p160 family of coactivators, which includes steroid receptor coactivator-1 (SRC-1), SRC-2 and SRC-3, in steroid receptor action in the brain. The expression, regulation and function of these coactivators in steroid-dependent gene expression in both brain and behaviour are discussed.
Collapse
Affiliation(s)
- M J Tetel
- Neuroscience Program, Wellesley College, Wellesley, MA, USA
| | | |
Collapse
|
32
|
Amandusson Å, Blomqvist A. Estrogenic influences in pain processing. Front Neuroendocrinol 2013; 34:329-49. [PMID: 23817054 DOI: 10.1016/j.yfrne.2013.06.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 06/19/2013] [Accepted: 06/20/2013] [Indexed: 12/24/2022]
Abstract
Gonadal hormones not only play a pivotal role in reproductive behavior and sexual differentiation, they also contribute to thermoregulation, feeding, memory, neuronal survival, and the perception of somatosensory stimuli. Numerous studies on both animals and human subjects have also demonstrated the potential effects of gonadal hormones, such as estrogens, on pain transmission. These effects most likely involve multiple neuroanatomical circuits as well as diverse neurochemical systems and they therefore need to be evaluated specifically to determine the localization and intrinsic characteristics of the neurons engaged. The aim of this review is to summarize the morphological as well as biochemical evidence in support for gonadal hormone modulation of nociceptive processing, with particular focus on estrogens and spinal cord mechanisms.
Collapse
Affiliation(s)
- Åsa Amandusson
- Department of Clinical Neurophysiology, Uppsala University, 751 85 Uppsala, Sweden.
| | | |
Collapse
|
33
|
O'Connell LA, Ding JH, Hofmann HA. Sex differences and similarities in the neuroendocrine regulation of social behavior in an African cichlid fish. Horm Behav 2013; 64:468-76. [PMID: 23899762 DOI: 10.1016/j.yhbeh.2013.07.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 07/21/2013] [Indexed: 01/13/2023]
Abstract
An individual's position in a social hierarchy profoundly affects behavior and physiology through interactions with community members, yet little is known about how the brain contributes to status differences between and within the social states or sexes. We aimed to determine sex-specific attributes of social status by comparing circulating sex steroid hormones and neural gene expression of sex steroid receptors in dominant and subordinate male and female Astatotilapia burtoni, a highly social African cichlid fish. We found that testosterone and 17β-estradiol levels are higher in males regardless of status and dominant individuals regardless of sex. Progesterone was found to be higher in dominant individuals regardless of sex. Based on pharmacological manipulations in males and females, progesterone appears to be a common mechanism for promoting courtship in dominant individuals. We also examined expression of androgen receptors, estrogen receptor α, and the progesterone receptor in five brain regions that are important for social behavior. Most of the differences in brain sex steroid receptor expression were due to sex rather than status. Our results suggest that the parvocellular preoptic area is a core region for mediating sex differences through androgen and estrogen receptor expression, whereas the progesterone receptor may mediate sex and status behaviors in the putative homologs of the nucleus accumbens and ventromedial hypothalamus. Overall our results suggest sex differences and similarities in the regulation of social dominance by gonadal hormones and their receptors in the brain.
Collapse
Affiliation(s)
- Lauren A O'Connell
- Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX 78705, USA; Section of Integrative Biology, University of Texas at Austin, Austin, TX 78705, USA
| | | | | |
Collapse
|
34
|
Kundakovic M, Gudsnuk K, Franks B, Madrid J, Miller RL, Perera FP, Champagne FA. Sex-specific epigenetic disruption and behavioral changes following low-dose in utero bisphenol A exposure. Proc Natl Acad Sci U S A 2013; 110:9956-61. [PMID: 23716699 PMCID: PMC3683772 DOI: 10.1073/pnas.1214056110] [Citation(s) in RCA: 364] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Bisphenol A (BPA) is an estrogenic endocrine disruptor widely used in the production of plastics. Increasing evidence indicates that in utero BPA exposure affects sexual differentiation and behavior; however, the mechanisms underlying these effects are unknown. We hypothesized that BPA may disrupt epigenetic programming of gene expression in the brain. Here, we provide evidence that maternal exposure during pregnancy to environmentally relevant doses of BPA (2, 20, and 200 µg/kg/d) in mice induces sex-specific, dose-dependent (linear and curvilinear), and brain region-specific changes in expression of genes encoding estrogen receptors (ERs; ERα and ERβ) and estrogen-related receptor-γ in juvenile offspring. Concomitantly, BPA altered mRNA levels of epigenetic regulators DNA methyltransferase (DNMT) 1 and DNMT3A in the juvenile cortex and hypothalamus, paralleling changes in estrogen-related receptors. Importantly, changes in ERα and DNMT expression in the cortex (males) and hypothalamus (females) were associated with DNA methylation changes in the ERα gene. BPA exposure induced persistent, largely sex-specific effects on social and anxiety-like behavior, leading to disruption of sexually dimorphic behaviors. Although postnatal maternal care was altered in mothers treated with BPA during pregnancy, the effects of in utero BPA were not found to be mediated by maternal care. However, our data suggest that increased maternal care may partially attenuate the effects of in utero BPA on DNA methylation. Overall, we demonstrate that low-dose prenatal BPA exposure induces lasting epigenetic disruption in the brain that possibly underlie enduring effects of BPA on brain function and behavior, especially regarding sexually dimorphic phenotypes.
Collapse
Affiliation(s)
- Marija Kundakovic
- Department of Psychology, Columbia University, New York, NY 10027; and
| | - Kathryn Gudsnuk
- Department of Psychology, Columbia University, New York, NY 10027; and
| | - Becca Franks
- Department of Psychology, Columbia University, New York, NY 10027; and
| | - Jesus Madrid
- Department of Psychology, Columbia University, New York, NY 10027; and
| | - Rachel L. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032
| | - Frederica P. Perera
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032
| | | |
Collapse
|
35
|
Wang Y, Xu L, Pan Y, Wang Z, Zhang Z. Species differences in the immunoreactive expression of oxytocin, vasopressin, tyrosine hydroxylase and estrogen receptor alpha in the brain of Mongolian gerbils (Meriones unguiculatus) and Chinese striped hamsters (Cricetulus barabensis). PLoS One 2013; 8:e65807. [PMID: 23762431 PMCID: PMC3676338 DOI: 10.1371/journal.pone.0065807] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/28/2013] [Indexed: 01/21/2023] Open
Abstract
Species differences in neurochemical expression and activity in the brain may play an important role in species-specific patterns of social behavior. In the present study, we used immunoreactive (ir) labeling to compare the regional density of cells containing oxytocin (OT), vasopressin (AVP), tyrosine hydroxylase (TH), or estrogen receptor alpha (ERα) staining in the brains of social Mongolian gerbils (Meriones unguiculatus) and solitary Chinese striped hamsters (Cricetulus barabensis). Multiple region- and neurochemical-specific species differences were found. In the anterior hypothalamus (AH), Mongolian gerbils had higher densities of AVP-ir and ERα-ir cells than Chinese striped hamsters. In the lateral hypothalamus (LH), Mongolian gerbils also had higher densities of AVP-ir and TH-ir cells, but a lower density of OT-ir cells, than Chinese striped hamsters. Furthermore, in the anterior nucleus of the medial preoptic area (MPOAa), Mongolian gerbils had higher densities of OT-ir and AVP-ir cells than Chinese striped hamsters, and an opposite pattern was found in the posterior nucleus of the MPOA (MPOAp). Some sex differences were also observed. Females of both species had higher densities of TH-ir cells in the MPOAa and of OT-ir cells in the intermediate nucleus of the MPOA (MPOAi) than males. Given the role of these neurochemicals in social behaviors, our data provide additional evidence to support the notion that species-specific patterns of neurochemical expression in the brain may be involved in species differences in social behaviors associated with different life strategies.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Linxi Xu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Yongliang Pan
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, United States of America
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
- * E-mail:
| |
Collapse
|
36
|
Wang H, Meyer K, Korz V. Stress induced hippocampal mineralocorticoid and estrogen receptor β gene expression and long-term potentiation in male adult rats is sensitive to early-life stress experience. Psychoneuroendocrinology 2013; 38:250-62. [PMID: 22776422 DOI: 10.1016/j.psyneuen.2012.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 01/06/2023]
Abstract
Glucocorticoid hormones and their receptors have been identified to be involved in emotional and cognitive disorders in early stressed subjects during adulthood. However, the impact of other steroid hormones and receptors has been considered less. Especially, functional roles of estrogen and estrogen receptors in male subjects are largely unknown. Therefore, we measured hippocampal concentrations of 17β-estradiol, corticosterone and testosterone, as well as the gene expression of estrogen receptor α and β (ERα, β), androgen receptor (AR), glucocorticoid (GR) and mineralocorticoid (MR) receptors after stress in adulthood in maternally separated (MS+; at postnatal days 14-16 for 6h each day) and control (MS-) male rats. In vivo hippocampal long-term potentiation (LTP) serves as a cellular model of learning and memory formation. Population spike- (PSA) and the fEPSP-LTP within the dentate gyrus (DG) were reinforced by elevated-platform-stress (EP-stress) in MS- but not in MS+ rats. MR- and ERβ-mRNA were upregulated 1h after EP-stress in MS- but not in MS+ rats as compared to non-stressed littermates. Infusion of an MR antagonist before LTP induction blocked early- and late-PSA- and -fEPSP-LTP, whereas blockade of ERβ impaired only the late PSA-LTP. Application of a DNA methyltransferase (DNMT) inhibitor partly restored the LTP-reinforcement in MS+ rats, accompanied by a retrieval of ERβ- but not MR-mRNA upregulation. Basal ERβ gene promoter methylation was similar between groups, whereas MS+ and MS- rats showed different methylation patterns across CpG sites after EP-stress. These findings indicate a key role of ERβ in early-stress mediated emotionality and emotion-induced late-LTP in adult male rats via DNA methylation mechanisms.
Collapse
Affiliation(s)
- Han Wang
- Leibniz Institute for Neurobiology, Brenneckestrasse 6, D-39118 Magdeburg, Germany
| | | | | |
Collapse
|
37
|
Meyer K, Korz V. Age dependent differences in the regulation of hippocampal steroid hormones and receptor genes: relations to motivation and cognition in male rats. Horm Behav 2013; 63:376-84. [PMID: 23238103 DOI: 10.1016/j.yhbeh.2012.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 11/28/2012] [Accepted: 12/03/2012] [Indexed: 11/16/2022]
Abstract
Estrogen and estrogenic functions are age-dependently involved in the modulation of learning, memory and mood in female humans and animals. However, the investigation of estrogenic effects in males has been largely neglected. Therefore, we investigated the hippocampal gene expression of estrogen receptors α and β (ERα, β) in 8-week-old, 12-week-old and 24-week-old male rats. To control for possible interactions between the expression of the estrogen receptor genes and other learning-related steroid receptors, androgen receptors (AR), corticosterone-binding glucocorticoid receptors (GR) and mineralocorticoid receptors (MR) were also measured. Furthermore, the concentrations of the ligands 17β-estradiol, testosterone and corticosterone were measured. The spatial training was conducted in a hole-board. The 8-week-old rats exhibited higher levels of general activity and exploration during the training and performed best with respect to spatial learning and memory, whereas no difference was found between the 12-week-old and 24-week-old rats. The trained 8-week-old rats exhibited increased gene expression of ERα compared with the untrained rats in this age group as well as the trained 12-week-old and 24-week-old rats. The concentrations of estradiol and testosterone, however, were generally higher in the 24-week-old rats than in the 8-week-old and 12-week-old rats. The ERα mRNA concentrations correlated positively with behavior that indicate general learning motivation. These results suggest a specific role of ERα in the age-related differences in motivation and subsequent success in the task. Thus, estrogen and estrogenic functions may play a more prominent role in young male behavior and development than has been previously assumed.
Collapse
Affiliation(s)
- K Meyer
- Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany.
| | | |
Collapse
|
38
|
|
39
|
Kastenberger I, Lutsch C, Schwarzer C. Activation of the G-protein-coupled receptor GPR30 induces anxiogenic effects in mice, similar to oestradiol. Psychopharmacology (Berl) 2012; 221:527-35. [PMID: 22143579 PMCID: PMC3350630 DOI: 10.1007/s00213-011-2599-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 11/21/2011] [Indexed: 11/28/2022]
Abstract
RATIONALE The influence of ovarian hormones on behaviour is well accepted, and oestrogen replacement therapy has proven to be beneficial in several cases of menopausal mood disorders. However, there are also some adverse effects of such a therapy, like anxiety and dysphoria. In fact, some women feel better at low levels of oestrogen and worse when levels fluctuate. Still, it is unclear which receptors might mediate negative emotional effects. OBJECTIVES The aim of this study was to identify which oestrogen receptor(s) are capable of mediating negative emotional effects and, therefore, may represent candidates responsible for the adverse side effects observed in oestrogen replacement therapy. RESULTS We provide evidence from mouse behavioural tests that oestrogen-induced anxiogenic-like effects might be mediated, at least in part, by the G protein-coupled receptor GPR30. The short-term application of specific agonists against the alpha and beta oestrogen receptors did not result in marked behavioural changes. In contrast, the specific stimulation of GPR30 in male and ovariectomized female mice induced anxiogenic effects. The anxiogenic effects induced by the specific GPR30 agonist G-1 were comparable (and non-accumulative) to those observed after low doses of the general oestrogen receptor agonist 17b-oestradiol in male mice, thereby reflecting the behavioural changes observed in intact female mice during early pro-oestrus. CONCLUSIONS Our data suggest that GPR30 induces acute anxiogenic effects of oestrogen in rodents. It is tempting to speculate that a potential imbalance in the expression of the anxiolytic beta oestrogen receptor and the anxiogenic GPR30 may also be involved in the negative symptoms of oestrogen replacement therapy in humans.
Collapse
Affiliation(s)
- Iris Kastenberger
- Department of Pharmacology, Innsbruck Medical University, Peter-Mayr-Str. 1a, 6020 Innsbruck, Austria
| | - Christian Lutsch
- Department of Pharmacology, Innsbruck Medical University, Peter-Mayr-Str. 1a, 6020 Innsbruck, Austria
| | - Christoph Schwarzer
- Department of Pharmacology, Innsbruck Medical University, Peter-Mayr-Str. 1a, 6020 Innsbruck, Austria
| |
Collapse
|
40
|
The distribution of estrogen receptor β mRNA in male and female green anole lizards. Brain Res 2012; 1430:43-51. [DOI: 10.1016/j.brainres.2011.10.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 10/24/2011] [Accepted: 10/27/2011] [Indexed: 12/30/2022]
|
41
|
Pan Y, Xu L, Wang Z, Zhang Z. Expression of oestrogen receptor α in the brain of Brandt's voles (Lasiopodomys brandtii ): sex differences and variations during ovarian cycles. J Neuroendocrinol 2011; 23:926-32. [PMID: 21848648 DOI: 10.1111/j.1365-2826.2011.02210.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Oestrogen receptor (ER) α plays an important role in a variety of cognitive and behavioural functions. It has been shown that ERα expression in the brain is sexually dimorphic and is influenced by circulating oestrogen. In the present study, we mapped ERα-immunoreactive (-ir) cells in the forebrain of Brandt's voles (Lasiopodomys brandtii) to examine differences in ERα-ir expression between males and females and to reveal variations of ERα-ir expression during ovarian cycles in females. ERα-ir cells were found in many forebrain regions, including the lateral septum, bed nucleus of the stria terminalis, medial preoptic area (MPOA), anterior, arcuate and ventral medial (VMH) nuclei of the hypothalamus, as well as medial (MeA) and anterior cortical nuclei of the amygdala. Females had more ERα-ir cells in the VMH than males. Females during ovarian oestrus, but not di-oestrus or pro-oestrus, also had more ERα-ir cells in the MPOA than males. Furthermore, females in ovarian di-oestrus or oestrus had more ERα-ir cells in the MeA than males. Together, these data indicate that ERα expression in the brain of Brandt's voles is sexually dimorphic in specific brain areas. In addition, variations in the levels of circulating oestrogen during ovarian cycles can affect ERα expression in the female brain in a region-specific manner.
Collapse
Affiliation(s)
- Y Pan
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | | | | | | |
Collapse
|
42
|
Seredynski AL, Ball GF, Balthazart J, Charlier TD. Specific activation of estrogen receptor alpha and beta enhances male sexual behavior and neuroplasticity in male Japanese quail. PLoS One 2011; 6:e18627. [PMID: 21533185 PMCID: PMC3077394 DOI: 10.1371/journal.pone.0018627] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 03/07/2011] [Indexed: 01/01/2023] Open
Abstract
Two subtypes of estrogen receptors (ER), ERα and ERβ, have been identified in humans and numerous vertebrates, including the Japanese quail. We investigated in this species the specific role(s) of each receptor in the activation of male sexual behavior and the underlying estrogen-dependent neural plasticity. Castrated male Japanese quail received empty (CX) or testosterone-filled (T) implants or were daily injected with the ER general agonist diethylstilbestrol (DES), the ERα-specific agonist PPT, the ERβ-specific agonist DPN or the vehicle, propylene glycol. Three days after receiving the first treatment, subjects were alternatively tested for appetitive (rhythmic cloacal sphincter movements, RCSM) and consummatory aspects (copulatory behavior) of male sexual behavior. 24 hours after the last behavioral testing, brains were collected and analyzed for aromatase expression and vasotocinergic innervation in the medial preoptic nucleus. The expression of RCSM was activated by T and to a lesser extent by DES and PPT but not by the ERβagonist DPN. In parallel, T fully restored the complete sequence of copulation, DES was partially active and the specific activation of ERα or ERβ only resulted in a very low frequency of mount attempts in few subjects. T increased the volume of the medial preoptic nucleus as measured by the dense cluster of aromatase-immunoreactive cells and the density of the vasotocinergic innervation within this nucleus. DES had only a weak action on vasotocinergic fibers and the two specific ER agonists did not affect these neural responses. Simultaneous activation of both receptors or treatments with higher doses may be required to fully activate sexual behavior and the associated neurochemical events.
Collapse
Affiliation(s)
- Aurore L. Seredynski
- Research Group in Behavioral Neuroendocrinology, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Gregory F. Ball
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jacques Balthazart
- Research Group in Behavioral Neuroendocrinology, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | - Thierry D. Charlier
- Research Group in Behavioral Neuroendocrinology, GIGA-Neurosciences, University of Liège, Liège, Belgium
- * E-mail:
| |
Collapse
|
43
|
Pfaff D, Waters E, Khan Q, Zhang X, Numan M. Minireview: estrogen receptor-initiated mechanisms causal to mammalian reproductive behaviors. Endocrinology 2011; 152:1209-17. [PMID: 21325045 PMCID: PMC3060638 DOI: 10.1210/en.2010-1007] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
While estrogen-facilitated changes in gene expression constitute some of the best-analyzed biochemical phenomena in the regulation of transcription, there have been at least two aspects of this topic that have led to much experimental work about estrogen actions on brain and behavior. The first task has required parsing various behavioral and neurochemical functions according to whether they depend on estrogen receptor-α or estrogen receptor-β. The second task has been the formulation of how nuclear actions of estrogens comport with membrane-initiated actions. With respect to these issues, applications of molecular endocrine approaches to lordosis behavior came first. Currently, the last in the chain of reproductive behaviors, maternal behavior, and an entire range of neural and cognitive functions even more complex in their determinants, must be analyzed using current molecular techniques. This minireview of estrogen actions on the chain of female reproductive behaviors highlights challenging new questions about estrogen actions on cells in the brain, questions that have important practical applications far beyond traditionally studied sex behaviors.
Collapse
Affiliation(s)
- Donald Pfaff
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, New York 10021, USA.
| | | | | | | | | |
Collapse
|
44
|
Waters EM, Yildirim M, Janssen WGM, Lou WYW, McEwen BS, Morrison JH, Milner TA. Estrogen and aging affect the synaptic distribution of estrogen receptor β-immunoreactivity in the CA1 region of female rat hippocampus. Brain Res 2011; 1379:86-97. [PMID: 20875808 PMCID: PMC3046233 DOI: 10.1016/j.brainres.2010.09.069] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 09/17/2010] [Accepted: 09/17/2010] [Indexed: 11/29/2022]
Abstract
Estradiol (E) mediates increased synaptogenesis in the hippocampal CA1 stratum radiatum (sr) and enhances memory in young and some aged female rats, depending on dose and age. Young female rats express more estrogen receptor α (ERα) immunolabeling in CA1sr spine synapse complexes than aged rats and ERα regulation is E sensitive in young but not aged rats. The current study examined whether estrogen receptor β (ERβ) expression in spine synapse complexes may be altered by age or E treatment. Young (3-4 months) and aged (22-23 months) female rats were ovariectomized 7 days prior to implantation of silastic capsules containing either vehicle (cholesterol) or E (10% in cholesterol) for 2 days. ERβ immunoreactivity (ir) in CA1sr was quantitatively analyzed using post-embedding electron microscopy. ERβ-ir was more prominent post-synaptically than pre-synaptically and both age and E treatment affected its synaptic distribution. While age decreased the spine synaptic complex localization of ERβ-ir (i.e., within 60 nm of the pre- and post-synaptic membranes), E treatment increased synaptic ERβ in both young and aged rats. In addition, the E treatment, but not age, increased dendritic shaft labeling. This data demonstrates that like ERα the levels of ERβ-ir decrease in CA1 axospinous synapses with age, however, unlike ERα the levels of ERβ-ir increase in these synapses in both young and aged rats in response to E. This suggests that synaptic ERβ may be a more responsive target to E, particularly in aged females.
Collapse
Affiliation(s)
- Elizabeth M Waters
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Tognoni CM, Chadwick JG, Ackeifi CA, Tetel MJ. Nuclear receptor coactivators are coexpressed with steroid receptors and regulated by estradiol in mouse brain. Neuroendocrinology 2011; 94:49-57. [PMID: 21311177 PMCID: PMC3150972 DOI: 10.1159/000323780] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 12/18/2010] [Indexed: 12/31/2022]
Abstract
BACKGROUND/AIMS The steroid hormones, including estradiol (E) and progesterone, act in the brain to regulate female reproductive behavior and physiology. These hormones mediate many of their biological effects by binding to their respective intracellular receptors. The receptors for estrogens (ER) and progestins (PR) interact with nuclear receptor coactivators to initiate transcription of steroid-responsive genes. Work from our laboratory and others reveals that nuclear receptor coactivators, including steroid receptor coactivator-1 (SRC-1) and SRC-2, function in brain to modulate ER-mediated induction of the PR gene and hormone-dependent behaviors. In order for steroid receptors and coactivators to function together, both must be expressed in the same cells. METHODS Triple-label immunofluorescence was used to determine if E-induced PR cells also express SRC-1 or SRC-2 in reproductively relevant brain regions of the female mouse. RESULTS The majority of E-induced PR cells in the medial preoptic area (61%), ventromedial nucleus of the hypothalamus (63%) and arcuate nucleus (76%) coexpressed both SRC-1 and SRC-2. A smaller proportion of PR cells expressed either SRC-1 or SRC-2, while a few PR cells expressed neither coactivator. In addition, compared to control animals, 17β-estradiol benzoate (EB) treatment increased SRC-1 levels in the arcuate nucleus, but not the medial preoptic area or the ventromedial nucleus of the hypothalamus. EB did not alter SRC-2 expression in any of the three brain regions analyzed. CONCLUSIONS Taken together, the present findings identify a population of cells in which steroid receptors and nuclear receptor coactivators may interact to modulate steroid sensitivity in brain and regulate hormone-dependent behaviors in female mice. Given that cell culture studies reveal that SRC-1 and SRC-2 can mediate distinct steroid-signaling pathways, the present findings suggest that steroids can produce a variety of complex responses in these specialized brain cells.
Collapse
Affiliation(s)
| | | | | | - Marc J. Tetel
- *Marc J. Tetel, Neuroscience Program, Wellesley College, 106 Central St., Wellesley, MA 02481 (USA), Tel. +1 781 283 3003, E-Mail
| |
Collapse
|
46
|
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 2010; 1379:119-36. [PMID: 21130078 DOI: 10.1016/j.brainres.2010.11.081] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [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
| | | | | | | | | |
Collapse
|
47
|
Fernández-Guasti A, Arteaga-López P, Antonio-Cabrera E. Copulation modifies AR and ERα mRNA expression in the male rat brain. Physiol Behav 2010; 101:738-45. [PMID: 20804777 DOI: 10.1016/j.physbeh.2010.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 08/13/2010] [Accepted: 08/23/2010] [Indexed: 01/03/2023]
Abstract
BACKGROUND/AIMS One day after male sexual behavior [one ejaculation or copulation to satiety (ad libitum copulation during 4h with the same female)] androgen receptor immunoreactivity (AR-ir) is decreased and estrogen receptor alpha immunoreactivity (ERα-ir) increased in various brain areas related with its control. Seven days after sexual satiety there was a limited recovery of sexual behavior accompanied by a partial recuperation in the AR-ir. In this study we evaluated if these changes in AR-ir and ERα-ir were paralleled by variations in their respective mRNA. METHODS Sexually experienced male rats were sacrificed at different intervals: immediately, 24h or seven days after sexual satiety or 24h after one ejaculation. The changes in AR and ERα mRNA were analyzed by in situ hybridization using digoxigenine-labeled oligonucleotide probes in the MPOA, LSV and the bed nucleus of the stria terminalis, medial division, anterior (BSTMA). RESULTS AR mRNA density was decreased in the MPOA and the LSV immediately and 24h after one ejaculation or sexual satiety. Seven days after copulating to satiety, there was a recovery of AR mRNA. In the BSTMA the different behavioral conditions did not modify the AR mRNA expression. In the MPOA, LSV and BSTMA the ERα mRNA increased after a single ejaculation and at all intervals after sexual satiety. CONCLUSION In some brain areas and after some intervals of sexual activity, the changes in steroid protein receptors expression seem to be consequence of parallel changes in the expression of the respective mRNA.
Collapse
Affiliation(s)
- Alonso Fernández-Guasti
- Department of Pharmacobiology, CINVESTAV, Calzada de los Tenorios 235, Col. Granjas Coapa, Del. Tlalpan, México City, D. F. 14330 México.
| | | | | |
Collapse
|