1
|
Peart DR, Andrade AK, Logan CN, Knackstedt LA, Murray JE. Regulation of Cocaine-related Behaviors by Estrogen and Progesterone. Neurosci Biobehav Rev 2022; 135:104584. [DOI: 10.1016/j.neubiorev.2022.104584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/30/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
|
2
|
Pang H, Jiang Y, Li J, Wang Y, Nie M, Xiao N, Wang S, Song Z, Ji F, Chang Y, Zheng Y, Yao K, Yao L, Li S, Li P, Song L, Lan X, Xu Z, Hu Z. Aberrant NAD + metabolism underlies Zika virus-induced microcephaly. Nat Metab 2021; 3:1109-1124. [PMID: 34385701 DOI: 10.1038/s42255-021-00437-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 07/07/2021] [Indexed: 12/18/2022]
Abstract
Zika virus (ZIKV) infection during pregnancy can cause microcephaly in newborns, yet the underlying mechanisms remain largely unexplored. Here, we reveal extensive and large-scale metabolic reprogramming events in ZIKV-infected mouse brains by performing a multi-omics study comprising transcriptomics, proteomics, phosphoproteomics and metabolomics approaches. Our proteomics and metabolomics analyses uncover dramatic alteration of nicotinamide adenine dinucleotide (NAD+)-related metabolic pathways, including oxidative phosphorylation, TCA cycle and tryptophan metabolism. Phosphoproteomics analysis indicates that MAPK and cyclic GMP-protein kinase G signaling may be associated with ZIKV-induced microcephaly. Notably, we demonstrate the utility of our rich multi-omics datasets with follow-up in vivo experiments, which confirm that boosting NAD+ by NAD+ or nicotinamide riboside supplementation alleviates cell death and increases cortex thickness in ZIKV-infected mouse brains. Nicotinamide riboside supplementation increases the brain and body weight as well as improves the survival in ZIKV-infected mice. Our study provides a comprehensive resource of biological data to support future investigations of ZIKV-induced microcephaly and demonstrates that metabolic alterations can be potentially exploited for developing therapeutic strategies.
Collapse
Affiliation(s)
- Huanhuan Pang
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
| | - Yisheng Jiang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Li
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Yushen Wang
- School of Life Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences (the PHOENIX Center), Beijing, China
| | - Meng Nie
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
| | - Nan Xiao
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
| | - Shuo Wang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhihong Song
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Fansen Ji
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Yafei Chang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Zheng
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Ke Yao
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
| | - LiAng Yao
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
| | - Shao Li
- Institute of TCM-X, MOE Key Laboratory of Bioinformatics / Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, China
| | - Peng Li
- School of Life Sciences, Tsinghua University, Beijing, China
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
- Shanghai Qi Zhi Institute, Shanghai, China
| | - Lei Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences (the PHOENIX Center), Beijing, China.
| | - Xun Lan
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China.
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China.
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China.
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.
| |
Collapse
|
3
|
Tibolone regulates systemic metabolism and the expression of sex hormone receptors in the central nervous system of ovariectomised rats fed with high-fat and high-fructose diet. Brain Res 2020; 1748:147096. [DOI: 10.1016/j.brainres.2020.147096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 02/04/2023]
|
4
|
Li X, Li C, Xu Y, Yao R, Li H, Ni W, Quan R, Zhang M, Liu L, Yu S, Ullah Y, Hu R, Li Y, Guo T, Wang X, Hu S. Analysis of pituitary transcriptomics indicates that lncRNAs are involved in the regulation of sheep estrus. Funct Integr Genomics 2020; 20:563-573. [PMID: 32114660 DOI: 10.1007/s10142-020-00735-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 11/17/2019] [Accepted: 02/06/2020] [Indexed: 12/13/2022]
Abstract
Seasonal estrus is a key factor limiting animal fertility, and understanding the molecular mechanisms that regulate animal estrus is important for improving animal fertility. The pituitary gland, which is the most important endocrine gland in mammals, plays an important role in regulating the physiological processes such as growth, development, and reproduction of animals. Here, we used RNA-seq technology to study the expression profile of lncRNAs in the anterior pituitary of sheep during estrus and anestrus. In this study, we identified a total of 995 lncRNAs, of which 335 lncRNAs were differentially expressed in two states (including 38 up-regulated and 297 down-regulated lncRNAs). RT-qPCR verified the expression levels of several lncRNAs. Target predictive analysis revealed that these lncRNAs can act in cis or trans and regulate the expression of genes involved in the regulation of sheep estrus. Target gene enrichment analysis of differentially expressed lncRNAs indicates that these lncRNAs can regulate sheep estrus by regulating hormone metabolism and energy metabolism. Through our research, we provide the expression profile of lncRNAs in the pituitary of sheep, which provides a valuable resource for further understanding of the genetic regulation of seasonal estrus in sheep from the perspective of lncRNAs.
Collapse
Affiliation(s)
- Xiaoyue Li
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Cunyuan Li
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China.,College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Yueren Xu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Rui Yao
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Huixiang Li
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Wei Ni
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China.
| | - Renzhe Quan
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Mengdan Zhang
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Li Liu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Shuting Yu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Yaseen Ullah
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Ruirui Hu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Yaxin Li
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Tao Guo
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Xiaokui Wang
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China
| | - Shengwei Hu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang, China.
| |
Collapse
|
5
|
Shevchouk OT, Ghorbanpoor S, Smith E, Liere P, Schumacher M, Ball GF, Cornil CA, Balthazart J. Behavioral evidence for sex steroids hypersensitivity in castrated male canaries. Horm Behav 2018; 103:80-96. [PMID: 29909262 DOI: 10.1016/j.yhbeh.2018.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 06/05/2018] [Accepted: 06/10/2018] [Indexed: 12/30/2022]
Abstract
In seasonally breeding songbirds such as canaries, singing behavior is predominantly under the control of testosterone and its metabolites. Short daylengths in the fall that break photorefractoriness are followed by increasing daylengths in spring that activate singing via both photoperiodic and hormonal mechanisms. However, we observed in a group of castrated male Fife fancy canaries maintained for a long duration under a short day photoperiod a large proportion of subjects that sang at high rates. This singing rate was not correlated with variation in the low circulating concentrations of testosterone. Treatment of these actively singing castrated male canaries with a combination of an aromatase inhibitor (ATD) and an androgen receptor blocker (flutamide) only marginally decreased this singing activity as compared to control untreated birds and did not affect various measures of song quality. The volumes of HVC and of the medial preoptic nucleus (POM) were also unaffected by these treatments but were relatively large and similar to volumes in testosterone-treated males. In contrast, peripheral androgen-sensitive structures such as the cloacal protuberance and syrinx mass were small, similar to what is observed in castrates. Together these data suggest that after a long-term steroid deprivation singing behavior can be activated by very low concentrations of testosterone. Singing normally depends on the activation by testosterone and its metabolites of multiple downstream neurochemical systems such as catecholamines, nonapeptides or opioids. These transmitter systems might become hypersensitive to steroid action after long term castration as they probably are at the end of winter during the annual cycle in seasonally breeding temperate zone species.
Collapse
Affiliation(s)
| | | | - Ed Smith
- Department of Psychology, University of Maryland, College Park, MD 20742, United States of America
| | - Philippe Liere
- INSERM UMR 1195 and Université Paris Sud and University Paris-Saclay, Le Kremlin-Bicêtre Cedex, France
| | - Michael Schumacher
- INSERM UMR 1195 and Université Paris Sud and University Paris-Saclay, Le Kremlin-Bicêtre Cedex, France
| | - Gregory F Ball
- Department of Psychology, University of Maryland, College Park, MD 20742, United States of America
| | | | | |
Collapse
|
6
|
Chu C, Deng J, Liu L, Cao Y, Wei X, Li J, Man Y. Nanoparticles combined with growth factors: recent progress and applications. RSC Adv 2016. [DOI: 10.1039/c6ra13636b] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Increasing attention has been focused on the applications of nanoparticles combined with growth factors (NPs/GFs) due to the substantial functions of GFs in regenerative medicine and disease treatments.
Collapse
Affiliation(s)
- Chenyu Chu
- State Key Laboratory of Oral Diseases
- West China Hospital of Stomatology
- Sichuan University
- Chengdu 610041
- China
| | - Jia Deng
- State Key Laboratory of Oral Diseases
- West China Hospital of Stomatology
- Sichuan University
- Chengdu 610041
- China
| | - Li Liu
- State Key Laboratory of Biotherapy and Laboratory for Aging Research
- West China Hospital
- Sichuan University and Collaborative Innovation Center for Biotherapy
- Chengdu
- China
| | - Yubin Cao
- State Key Laboratory of Oral Diseases
- West China Hospital of Stomatology
- Sichuan University
- Chengdu 610041
- China
| | - Xiawei Wei
- State Key Laboratory of Biotherapy and Laboratory for Aging Research
- West China Hospital
- Sichuan University and Collaborative Innovation Center for Biotherapy
- Chengdu
- China
| | - Jidong Li
- Research Center for Nano Biomaterials
- Analytical & Testing Center
- Sichuan University
- Chengdu 610041
- P. R. China
| | - Yi Man
- State Key Laboratory of Oral Diseases
- West China Hospital of Stomatology
- Sichuan University
- Chengdu 610041
- China
| |
Collapse
|
7
|
Zhang D, Jiang S, Meng H. Role of the Insulin-Like Growth Factor Type 1 Receptor in the Pathogenesis of Diabetic Encephalopathy. Int J Endocrinol 2015; 2015:626019. [PMID: 26089889 PMCID: PMC4451562 DOI: 10.1155/2015/626019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022] Open
Abstract
Defective cognitive function is common in patients with diabetes, suggesting that insulin normally exerts anabolic actions in neuron, namely, diabetic encephalopathy. However, because insulin can cross-activate the insulin-like growth factor type 1 receptor (IGF-1R), which also functions in most of tissues, such as muscle and bone, it has been difficult to establish the direct (IGF-1-independent) actions of insulin in the pathogenesis of diabetic encephalopathy. To overcome this problem, we examined insulin signaling and action in primary PC-12 cells engineered for conditional disruption of the IGF-1 receptor (ΔIGF-1R). The results showed that the lower glucose metabolism and high expression of IGF-1R occurred in the brain of the DE rat model. The results also showed the defect of IGF-1R could significantly improve the ability of glucose consumption and enhance sensitivity to insulin-induced IR and Akt phosphorylation in PC12 cells. And meanwhile, IGF-1R allele gene knockout (IGF-1R(neo)) mice treated with HFD/STZ had better cognitive abilities than those of wild mice. Those results indicate that insulin exerts direct anabolic actions in neuron-like cells by activation of its cognate receptor and prove that IGF-1R plays an important role in the pathogenesis of diabetic encephalopathy.
Collapse
Affiliation(s)
- Duo Zhang
- Department of Radiology, Affiliated Hospital of BeiHua University, JiLin 132011, China
| | - Shuang Jiang
- College of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Heng Meng
- Department of Radiology, Affiliated Hospital of BeiHua University, JiLin 132011, China
- *Heng Meng:
| |
Collapse
|
8
|
Fortress AM, Frick KM. Epigenetic regulation of estrogen-dependent memory. Front Neuroendocrinol 2014; 35:530-49. [PMID: 24878494 PMCID: PMC4174980 DOI: 10.1016/j.yfrne.2014.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 02/09/2023]
Abstract
Hippocampal memory formation is highly regulated by post-translational histone modifications and DNA methylation. Accordingly, these epigenetic processes play a major role in the effects of modulatory factors, such as sex steroid hormones, on hippocampal memory. Our laboratory recently demonstrated that the ability of the potent estrogen 17β-estradiol (E2) to enhance hippocampal-dependent novel object recognition memory in ovariectomized female mice requires ERK-dependent histone H3 acetylation and DNA methylation in the dorsal hippocampus. Although these data provide valuable insight into the chromatin modifications that mediate the memory-enhancing effects of E2, epigenetic regulation of gene expression is enormously complex. Therefore, more research is needed to fully understand how E2 and other hormones employ epigenetic alterations to shape behavior. This review discusses the epigenetic alterations shown thus far to regulate hippocampal memory, briefly reviews the effects of E2 on hippocampal function, and describes in detail our work on epigenetic regulation of estrogenic memory enhancement.
Collapse
Affiliation(s)
- Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
9
|
Frye C, Koonce C, Walf A. Role of pregnane xenobiotic receptor in the midbrain ventral tegmental area for estradiol- and 3α,5α-THP-facilitated lordosis of female rats. Psychopharmacology (Berl) 2014; 231:3365-74. [PMID: 24435323 PMCID: PMC4102666 DOI: 10.1007/s00213-013-3406-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 12/05/2013] [Indexed: 12/17/2022]
Abstract
RATIONALE Progesterone and its metabolite, 5α-pregnan-3α-ol-20-one (3α,5α-THP), have actions in the ventral tegmental area (VTA) that are required for lordosis, a characteristic mating posture of female rodents. 17β-estradiol (estradiol) co-varies with progestogens over natural cycles, enhances production of 3α,5α-THP, and is required for successful reproductive behavior. OBJECTIVES A question of interest is the role of pregnane xenobiotic receptor (PXR), a nuclear receptor that regulates enzymes needed for the production of 3α,5α-THP, for estradiol-mediated lordosis. The hypothesis tested was that if PXR is involved in estradiol-mediated biosynthesis of 3α,5α-THP and reproductive behavior, knocking down expression of PXR in the VTA of estradiol-primed, but not vehicle-primed, rats should decrease lordosis and midbrain 3α,5α-THP; effects may be attenuated by 3α,5α-THP administered to the VTA. METHODS Ovariectomized rats were administered subcutaneous injections of oil vehicle or estradiol. Rats were then administered PXR antisense oligonucleotides (PXR AS-ODNs; which are expected to locally knock down expression of PXR), or control (saline), infusions to the VTA. Rats were administered 3α,5α-THP or vehicle via infusions to the VTA. Reproductive behavior (paced mating task) of rats was determined in addition to exploratory (open field), affective (elevated plus maze), and pro-social (social interaction task) behavior. RESULTS Reproductive behavior (i.e., increased lordosis) was enhanced with estradiol-priming and infusions of 3α,5α-THP to the VTA. Infusions of PXR AS-ODNs to the VTA attenuated responses in estradiol-, but not vehicle-, primed rats, compared to control infusions. CONCLUSIONS PXR may be involved in a neuroregulatory response involving biosynthesis of 3α,5α-THP in the midbrain VTA of estradiol-primed rats.
Collapse
Affiliation(s)
- C.A. Frye
- Dept. of Psychology, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,Dept. of Biological Sciences, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,The Centers for Neuroscience, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,The Centers for Life Sciences Research, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,Department of Chemistry, Institute for Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775,IDeA Network of Biomedical Excellence, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775
| | - C.J. Koonce
- Dept. of Psychology, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,Department of Chemistry, Institute for Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775
| | - A.A. Walf
- Dept. of Psychology, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,Department of Chemistry, Institute for Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775,IDeA Network of Biomedical Excellence, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775
| |
Collapse
|
10
|
Schumacher M, Mattern C, Ghoumari A, Oudinet JP, Liere P, Labombarda F, Sitruk-Ware R, De Nicola AF, Guennoun R. Revisiting the roles of progesterone and allopregnanolone in the nervous system: resurgence of the progesterone receptors. Prog Neurobiol 2013; 113:6-39. [PMID: 24172649 DOI: 10.1016/j.pneurobio.2013.09.004] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/15/2013] [Accepted: 09/21/2013] [Indexed: 02/08/2023]
Abstract
Progesterone is commonly considered as a female reproductive hormone and is well-known for its role in pregnancy. It is less well appreciated that progesterone and its metabolite allopregnanolone are also male hormones, as they are produced in both sexes by the adrenal glands. In addition, they are synthesized within the nervous system. Progesterone and allopregnanolone are associated with adaptation to stress, and increased production of progesterone within the brain may be part of the response of neural cells to injury. Progesterone receptors (PR) are widely distributed throughout the brain, but their study has been mainly limited to the hypothalamus and reproductive functions, and the extra-hypothalamic receptors have been neglected. This lack of information about brain functions of PR is unexpected, as the protective and trophic effects of progesterone are much investigated, and as the therapeutic potential of progesterone as a neuroprotective and promyelinating agent is currently being assessed in clinical trials. The little attention devoted to the brain functions of PR may relate to the widely accepted assumption that non-reproductive actions of progesterone may be mainly mediated by allopregnanolone, which does not bind to PR, but acts as a potent positive modulator of γ-aminobutyric acid type A (GABA(A) receptors. The aim of this review is to critically discuss effects of progesterone on the nervous system via PR, and of allopregnanolone via its modulation of GABA(A) receptors, with main focus on the brain.
Collapse
Affiliation(s)
- M Schumacher
- UMR 788 Inserm and University Paris-Sud, Kremlin-Bicêtre, France.
| | - C Mattern
- M et P Pharma AG, Emmetten, Switzerland
| | - A Ghoumari
- UMR 788 Inserm and University Paris-Sud, Kremlin-Bicêtre, France
| | - J P Oudinet
- UMR 788 Inserm and University Paris-Sud, Kremlin-Bicêtre, France
| | - P Liere
- UMR 788 Inserm and University Paris-Sud, Kremlin-Bicêtre, France
| | - F Labombarda
- Instituto de Biologia y Medicina Experimental and University of Buenos Aires, Argentina
| | - R Sitruk-Ware
- Population Council and Rockefeller University, New York, USA
| | - A F De Nicola
- Instituto de Biologia y Medicina Experimental and University of Buenos Aires, Argentina
| | - R Guennoun
- UMR 788 Inserm and University Paris-Sud, Kremlin-Bicêtre, France
| |
Collapse
|
11
|
Effects of noradrenergic alpha-2 receptor antagonism or noradrenergic lesions in the ventral bed nucleus of the stria terminalis and medial preoptic area on maternal care in female rats. Psychopharmacology (Berl) 2012; 224:263-76. [PMID: 22644129 PMCID: PMC3652389 DOI: 10.1007/s00213-012-2749-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 05/12/2012] [Indexed: 12/22/2022]
Abstract
RATIONALE Maternal behavior in laboratory rats requires a network of brain structures including the ventral bed nucleus of the stria terminalis (BSTv) and medial preoptic area (mPOA). Neurotransmitter systems in the BSTv and mPOA influencing maternal behaviors are not well understood, although norepinephrine is an excellent candidate because the BSTv contains the densest noradrenergic fiber plexus in the forebrain and norepinephrine in the mPOA is known to influence other female reproductive functions. OBJECTIVES We hypothesized that downregulated noradrenergic activity in the BSTv and mPOA is necessary for mothering. METHODS Postpartum mother-litter interactions were observed after BSTv infusion of yohimbine (an α2 autoreceptor antagonist that increases norepinephrine release), and after BSTv or mPOA infusion of the more selective α2 autoreceptor antagonist idazoxan. Lastly, noradrenergic input to the BSTv/mPOA was selectively lesioned in nulliparous rats with anti-DBH-saporin to determine if this would facilitate mothering. RESULTS BSTv yohimbine almost abolished retrieval of pups but did not significantly affect dams' ability to initiate contact, lick, or nurse them. BSTv idazoxan disrupted retrieval somewhat less than yohimbine, but significantly reduced nursing. mPOA idazoxan impaired retrieval more severely than that found after BSTv infusion. Anti-DBH-saporin almost eliminated noradrenergic terminals in the BSTv and reduced them by over 60% in the mPOA, but did not promote maternal responding. It also did not affect females' anxiety-related behavior. CONCLUSIONS Downregulated noradrenergic activity in the BSTv and mPOA is necessary for postpartum maternal behavior in rats, but eliminating this system alone is insufficient to promote maternal behaviors in nulliparous females.
Collapse
|
12
|
Uphouse L, Adams S, Miryala CSJ, Hassell J, Hiegel C. RU486 blocks effects of allopregnanolone on the response to restraint stress. Pharmacol Biochem Behav 2012; 103:568-72. [PMID: 23046854 DOI: 10.1016/j.pbb.2012.09.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 09/11/2012] [Accepted: 09/30/2012] [Indexed: 11/28/2022]
Abstract
These experiments were designed to provide information about the potential involvement of progesterone receptors in the ability of allopregnanolone (3α-hydroxy-5α-pregnan-20-one) to reduce the lordosis-inhibiting effects of restraint stress. Ovariectomized Fischer rats were hormonally primed with 10 μg estradiol benzoate and 4 mg/kg allopregnanolone or vehicle. One hour before allopregnanolone, rats were injected with the progesterone receptor antagonist, RU486 (11β-(4-dimethylamino)phenyl-17β-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one), or vehicle. Four hours after allopregnanolone or vehicle, sexual behavior was examined before and after a 5-min restraint stress. Lordosis behavior of rats primed only with estradiol benzoate declined after the 5 min of restraint while allopregnanolone prevented this decline. RU486 attenuated the ability of allopregnanolone to prevent the restraint-induced decline in lordosis behavior. These findings are consistent with earlier suggestions that progesterone receptors are involved in allopregnanolone's ability to reduce the effects of restraint stress.
Collapse
Affiliation(s)
- Lynda Uphouse
- Department of Biology, Texas Woman's University, United States.
| | | | | | | | | |
Collapse
|
13
|
Estrous behavior in dairy cows: identification of underlying mechanisms and gene functions. Animal 2012; 4:446-53. [PMID: 22443949 DOI: 10.1017/s1751731109991169] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Selection in dairy cattle for a higher milk yield has coincided with declined fertility. One of the factors is reduced expression of estrous behavior. Changes in systems that regulate the estrous behavior could be manifested by altered gene expression. This literature review describes the current knowledge on mechanisms and genes involved in the regulation of estrous behavior. The endocrinological regulation of the estrous cycle in dairy cows is well described. Estradiol (E2) is assumed to be the key regulator that synchronizes endocrine and behavioral events. Other pivotal hormones are, for example, progesterone, gonadotropin releasing hormone and insulin-like growth factor-1. Interactions between the latter and E2 may play a role in the unfavorable effects of milk yield-related metabolic stress on fertility in high milk-producing dairy cows. However, a clear understanding of how endocrine mechanisms are tied to estrous behavior in cows is only starting to emerge. Recent studies on gene expression and signaling pathways in rodents and other animals contribute to our understanding of genes and mechanisms involved in estrous behavior. Studies in rodents, for example, show that estrogen-induced gene expression in specific brain areas such as the hypothalamus play an important role. Through these estrogen-induced gene expressions, E2 alters the functioning of neuronal networks that underlie estrous behavior, by affecting dendritic connections between cells, receptor populations and neurotransmitter releases. To improve the understanding of complex biological networks, like estrus regulation, and to deal with the increasing amount of genomic information that becomes available, mathematical models can be helpful. Systems biology combines physiological and genomic data with mathematical modeling. Possible applications of systems biology approaches in the field of female fertility and estrous behavior are discussed.
Collapse
|
14
|
Cornil CA, Ball GF, Balthazart J. Rapid control of male typical behaviors by brain-derived estrogens. Front Neuroendocrinol 2012; 33:425-46. [PMID: 22983088 PMCID: PMC3496013 DOI: 10.1016/j.yfrne.2012.08.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/13/2012] [Accepted: 08/17/2012] [Indexed: 01/01/2023]
Abstract
Beside their genomic mode of action, estrogens also activate a variety of cellular signaling pathways through non-genomic mechanisms. Until recently, little was known regarding the functional significance of such actions in males and the mechanisms that control local estrogen concentration with a spatial and time resolution compatible with these non-genomic actions had rarely been examined. Here, we review evidence that estrogens rapidly modulate a variety of behaviors in male vertebrates. Then, we present in vitro work supporting the existence of a control mechanism of local brain estrogen synthesis by aromatase along with in vivo evidence that rapid changes in aromatase activity also occur in a region-specific manner in response to changes in the social or environmental context. Finally, we suggest that the brain estrogen provision may also play a significant role in females. Together these data bolster the hypothesis that brain-derived estrogens should be considered as neuromodulators.
Collapse
Affiliation(s)
- Charlotte A Cornil
- GIGA Neurosciences, Research Group in Behavioral Neuroendocrinology, University of Liège, Liège, Belgium.
| | | | | |
Collapse
|
15
|
Mani SK, Oyola MG. Progesterone signaling mechanisms in brain and behavior. Front Endocrinol (Lausanne) 2012; 3:7. [PMID: 22649404 PMCID: PMC3355960 DOI: 10.3389/fendo.2012.00007] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Accepted: 01/10/2012] [Indexed: 11/25/2022] Open
Abstract
Steroid hormone, progesterone, modulates neuroendocrine functions in the central nervous system resulting in alterations in physiology and behavior. These neuronal effects are mediated primarily by intracellular progestin receptors (PRs) in the steroid-sensitive neurons, resulting in transcription-dependent genomic actions (classical mechanism). In addition to progesterone, intracellular PRs can also be activated in a "ligand-independent" manner by neurotransmitters, peptide growth factors, cyclic nucleotides, and neurosteroids. Recent studies indicate that rapid, non-classical progesterone actions involving cytoplasmic kinase signaling and/or extranuclear PRs can result in both transcription-independent and transcription-dependent actions. Cross-talk between extranuclear and classical intracellular signaling pathways promotes progesterone-dependent behavior in mammals. This review focuses on the mechanisms by which progesterone-initiated signaling mechanisms converge with PRs in the brain to modulate reproductive behavior in female rodents.
Collapse
Affiliation(s)
- Shaila K Mani
- Center on Addiction, Learning and Memory, Department of Neuroscience, Baylor College of Medicine Houston, TX, USA.
| | | |
Collapse
|
16
|
Panzica GC, Balthazart J, Frye CA, Garcia-Segura LM, Herbison AE, Mensah-Nyagan AG, McCarthy MM, Melcangi RC. Milestones on Steroids and the Nervous System: 10 years of basic and translational research. J Neuroendocrinol 2012; 24:1-15. [PMID: 22188420 DOI: 10.1111/j.1365-2826.2011.02265.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
During the last 10 years, the conference on 'Steroids and Nervous System' held in Torino (Italy) has been an important international point of discussion for scientists involved in this exciting and expanding research field. The present review aims to recapitulate the main topics that have been presented through the various meetings. Two broad areas have been explored: the impact of gonadal hormones on brain circuits and behaviour, as well as the mechanism of action of neuroactive steroids. Relationships among steroids, brain and behaviour, the sexual differentiation of the brain and the impact of gonadal hormones, the interactions of exogenous steroidal molecules (endocrine disrupters) with neural circuits and behaviour, and how gonadal steroids modulate the behaviour of gonadotrophin-releasing hormone neurones, have been the topics of several lectures and symposia during this series of meetings. At the same time, many contributions have been dedicated to the biosynthetic pathways, the physiopathological relevance of neurosteroids, the demonstration of the cellular localisation of different enzymes involved in neurosteroidogenesis, the mechanisms by which steroids may exert some of their effects, both the classical and nonclassical actions of different steroids, the role of neuroactive steroids on neurodegeneration, neuroprotection, and the response of the neural tissue to injury. In these 10 years, this field has significantly advanced and neuroactive steroids have emerged as new potential therapeutic tools to counteract neurodegenerative events.
Collapse
Affiliation(s)
- G C Panzica
- Laboratory of Neuroendocrinology, Department of Anatomy, Pharmacology and Forensic Medicine, Neuroscience Institute of Turin (NIT), University of Torino, Torino, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Sun Y, Todd BJ, Thornton K, Etgen AM, Neal-Perry G. Differential effects of hypothalamic IGF-I on gonadotropin releasing hormone neuronal activation during steroid-induced LH surges in young and middle-aged female rats. Endocrinology 2011; 152:4276-87. [PMID: 21914776 PMCID: PMC3199007 DOI: 10.1210/en.2011-1051] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Interactions between brain IGF-I receptors and estrogen receptors regulate female reproductive physiology and behavior. The present study investigated potential mechanisms by which IGF-I receptors in the neuroendocrine hypothalamus regulate GnRH neuronal activation and LH release in young and middle-aged female rats under estradiol (E2) positive feedback conditions. We infused vehicle, IGF-I, or JB-1, a selective antagonist of IGF-I receptors, into the third ventricle of ovariectomized female rats primed with E2 and progesterone or vehicle. In young females, blockade of IGF-I receptors attenuated the steroid hormone-induced LH surge, reduced the percent of GnRH neurons expressing c-fos on the day of the LH surge, and decreased the total number of neurons expressing c-fos in the preoptic area. Middle-aged females had fewer GnRH neurons expressing c-fos during the LH surge than young females, and the LH surge amplitude was attenuated. Infusion of an IGF-I dose previously shown to increase LH surge amplitude did not increase the percent of GnRH neurons expressing c-fos in middle-aged females. Brain IGF-I receptor blockade did not modify E2 induction of progestin receptor-immunoreactive neurons in the preoptic area, arcuate, or ventromedial hypothalamus of young rats. These findings indicate that brain IGF-I receptors are required for E2 activation of GnRH neurons in young rats and for robust GnRH release from axon terminals in middle-aged females. IGF-I likely exerts its effects by actions on E2-sensitive neurons that are upstream of GnRH neurons and terminals.
Collapse
Affiliation(s)
- Yan Sun
- Department of Obstetrics/Gynecology and Women's Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Ullmann 1211, Bronx, New York 10461, USA
| | | | | | | | | |
Collapse
|
18
|
Bonthuis PJ, Patteson JK, Rissman EF. Acquisition of sexual receptivity: roles of chromatin acetylation, estrogen receptor-alpha, and ovarian hormones. Endocrinology 2011; 152:3172-81. [PMID: 21652725 PMCID: PMC3138229 DOI: 10.1210/en.2010-1001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Sexually naïve, hormone-primed, C57BL/6J female mice are not receptive to mating attempts by conspecific males. Repeated experience with sexually active males and concurrent treatment with estradiol and progesterone gradually increases female receptivity over the course of five trials to maximal levels. Ovarian hormones activate their cognate nuclear steroid receptors estrogen receptor-α and progesterone receptor to induce female sexual receptivity. Nuclear receptors recruit coactivators of transcription that include histone acetyltransferases to hormone responsive genes. In this set of studies, we found that the histone deacetylase inhibitor sodium butyrate enhances the experiential acquisition of receptivity. Evidence is provided that the actions of sodium butyrate on receptivity require activated estrogen receptor-α and progesterone.
Collapse
Affiliation(s)
- Paul J Bonthuis
- Department of Biochemistry and Molecular Genetics and Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | |
Collapse
|
19
|
Frye CA, Paris JJ. Progesterone turnover to its 5α-reduced metabolites in the ventral tegmental area of the midbrain is essential for initiating social and affective behavior and progesterone metabolism in female rats. J Endocrinol Invest 2011; 34:e188-99. [PMID: 21060252 PMCID: PMC3376830 DOI: 10.3275/7334] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Among women and female rodents, progesterone (P) influences social affiliation and affect. These effects may be partly due to formation of its 5α-reduced, 3α- hydroxylated metabolite, 5α-pregnan-3α-ol-20-one (3α,5α- THP). AIM To elucidate whether actions of 3α,5α-THP in the midbrain ventral tegmental area (VTA) are both necessary and sufficient to enhance non-sexual and sexual social behaviors, affect, and central 3α,5α-THP metabolism. MATERIALS AND METHODS P and 3α,5α-THP formation were unperturbed or blocked in VTA via infusions of vehicle, PK11195 (400 ng), and/or indomethacin (10 μg). Rats then received subsequent infusions of vehicle or 3α,5α-THP (100 ng) and were assessed in a battery of tasks that included open field (exploration), elevated plus maze (anxiety behavior), social interaction (social affiliation), and paced mating (sexual behavior) or were not tested. Metabolic turnover of P to its 5α-reduced metabolites was assessed in plasma, midbrain, hippocampus, frontal cortex, diencephalon, and remaining subcortical tissues (control interbrain). RESULTS Infusions of any combination of inhibitors significantly reduced social and affective behavior in all tasks compared to vehicle, concomitant with reduced turnover of P to its 5α-reduced metabolites, in midbrain only. Subsequent infusions of 3α,5α-THP significantly reinstated/enhanced anti- anxiety behavior, lordosis, and P turnover to its 5α-reduced metabolites in midbrain, as well as hippocampus, cortex, and diencephalon (but not plasma or interbrain). CONCLUSIONS These data are the first to provide direct evidence that actions of 3α,5α-THP in the VTA are both necessary and sufficient for social and affective behavior, as well as initiation of central 5α-reduction.
Collapse
Affiliation(s)
- C A Frye
- Department of Psychology, The University at Albany-SUNY, Albany, USA.
| | | |
Collapse
|
20
|
Gómez-Camarillo MA, Beyer C, Lucio RA, García-Juárez M, González-Arenas A, Camacho-Arroyo I, Komisaruk BR, González-Flores O. Differential effects of progesterone and genital stimulation on sequential inhibition of estrous behavior and progesterone receptor expression in the rat brain. Brain Res Bull 2011; 85:201-6. [PMID: 21515343 DOI: 10.1016/j.brainresbull.2011.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2010] [Revised: 03/25/2011] [Accepted: 04/10/2011] [Indexed: 11/28/2022]
Abstract
The effect of genital stimulation, either by vaginocervical stimulation (VCS) using a calibrated vaginal probe combined with manual flank stimulation (FS), or by mounts performed by the male, on the hypothalamus and preoptic area concentration of the progesterone receptors A (PR-A) and B (PR-B) was assessed in ovariectomized (ovx) estrogen-primed rats. VCS/FS or stimulation provided by male mounts, even without intromission, significantly decreased PR-B concentration in the hypoythalamus. Down regulation of PR produced by genital stimulation was quantitatively similar to that elicited by progesterone (P) administration. Bilateral or unilateral transection of the pelvic or the pudendal nerves prevented down regulation elicited by VCS/FS. Repeated VCS/FS elicited lordosis behavior in most ovx estrogen primed rats, but the lordosis intensity was lower than that observed in response to P. P administered to ovx estrogen primed rats, induced sequential inhibition, i.e., failure to display estrous behavior in response to a second P injection (24h after the initial P injection). VCS/FS failed to elicit sequential inhibition, since rats responded with normal estrous behavior to the second injection of P. This suggests that down regulation by VCS, by contrast with P, failed to inhibit the subpopulation of PR involved in the facilitation of estrous behavior by P.
Collapse
Affiliation(s)
- Madaí A Gómez-Camarillo
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Plaza Hidalgo S/N, Panotla, Tlaxcala CP, 90140, México
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Shibuya H, Sakai K, Kabir-Salmani M, Wachi Y, Iwashita M. Polymerization of insulin-like growth factor-binding protein-1 (IGFBP-1) potentiates IGF-I actions in placenta. J Cell Physiol 2011; 226:434-9. [PMID: 20672288 DOI: 10.1002/jcp.22349] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Insulin-like growth factor (IGF)-binding protein-1 (IGFBP-1), the main secretory protein of decidua that binds to IGFs and has been shown to inhibit or stimulate IGFs' bioactivities. Polymerization, one of the posttranslational modifications of IGFBP-1, has been shown to lead to loss of inhibiting effect of IGFBP-1 on IGF-I actions. The current studies were undertaken to elucidate the effects of steroid hormones on IGFBP-1 polymerization in trophoblast cell cultures. Placental tissues were obtained during legal, elective procedures of termination of pregnancy performed between 7 and 10 weeks of gestation, and primary trophoblast cells were separated. IGFBP-1 polymerization was analyzed by SDS-PAGE and immunoblotting. IGFBP-1 was polymerized when IGFBP-1 was added to trophoblast cell cultures. Polymerization of IGFBP-1 was inhibited by the addition of anti-tissue transglutaminase antibody into the culture media. There was an increase in the intensity of polymerized IGFBP-1 bands with the addition of medroxyprogesterone acetate (MPA), while no such difference was observed upon treatment with estradiol. MPA also increased the expression of tissue transglutaminase on trophoblast cell membranes. IGF-I stimulated trophoblast cell migration, while IGFBP-1 inhibited this IGF-I-induced trophoblast response. Addition of MPA attenuated the inhibitory effects of IGFBP-1 on IGF-I-induced trophoblast cell migration. IGFBP-1 was polymerized by tissue transglutaminase on the cell surface of trophoblasts, and MPA increased tissue transglutaminase expression on the cell surface and facilitated IGFBP-1 polymerization. These results suggest that progesterone might facilitate polymerization of decidua-secreted IGFBP-1 and increase IGF-I actions at feto-maternal interface, thereby stimulating trophoblast invasion of maternal uterus.
Collapse
Affiliation(s)
- Hiromi Shibuya
- Department of Obstetrics and Gynecology, Kyorin University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
22
|
Roepke TA, Ronnekleiv OK, Kelly MJ. Physiological consequences of membrane-initiated estrogen signaling in the brain. Front Biosci (Landmark Ed) 2011; 16:1560-73. [PMID: 21196248 DOI: 10.2741/3805] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Many of the actions of 17beta-estradiol (E2) in the central nervous system (CNS) are mediated via the classical nuclear steroid receptors, ER(alpha) and ERbeta, which interact with the estrogen response element to modulate gene expression. In addition to the nuclear-initiated estrogen signaling, E2 signaling in the brain can occur rapidly within minutes prior to any sufficient effects on transcription of relevant genes. These rapid, membrane-initiated E2 signaling mechanisms have now been characterized in many brain regions, most importantly in neurons of the hypothalamus and hippocampus. Furthermore, our understanding of the physiological effects of membrane-initiated pathways is now a major field of interest in the hypothalamic control of reproduction, energy balance, thermoregulation and other homeostatic functions as well as the effects of E2 on physiological and pathophysiological functions of the hippocampus. Membrane signaling pathways impact neuronal excitability, signal transduction, cell death, neurotransmitter release and gene expression. This review will summarize recent findings on membrane-initiated E2 signaling in the hypothalamus and hippocampus and its contribution to the control of physiological and behavioral functions.
Collapse
Affiliation(s)
- Troy A Roepke
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | | | | |
Collapse
|
23
|
Activation of progestin receptors in female reproductive behavior: Interactions with neurotransmitters. Front Neuroendocrinol 2010; 31:157-71. [PMID: 20116396 PMCID: PMC2849835 DOI: 10.1016/j.yfrne.2010.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 01/20/2010] [Accepted: 01/21/2010] [Indexed: 01/22/2023]
Abstract
The steroid hormone, progesterone (P), modulates neuroendocrine functions in the central nervous system resulting in alterations in physiology and reproductive behavior in female mammals. A wide body of evidence indicates that these neural effects of P are predominantly mediated via their intracellular progestin receptors (PRs) functioning as "ligand-dependent" transcription factors in the steroid-sensitive neurons regulating genes and genomic networks. In addition to P, intracellular PRs can be activated by neurotransmitters, growth factors and cyclic nucleotides in a ligand-independent manner via crosstalk and convergence of pathways. Furthermore, recent studies indicate that rapid signaling events associated with membrane PRs and/or extra-nuclear, cytoplasmic PRs converge with classical PR activated pathways in neuroendocrine regulation of female reproductive behavior. The molecular mechanisms, by which multiple signaling pathways converge on PRs to modulate PR-dependent female reproductive behavior, are discussed in this review.
Collapse
|
24
|
Amorim MA, Guerra-Araiza C, Garcia-Segura LM. Progesterone as a regulator of phosphorylation in the central nervous system. Horm Mol Biol Clin Investig 2010; 4:601-7. [DOI: 10.1515/hmbci.2010.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/05/2010] [Indexed: 11/15/2022]
Abstract
AbstractProgesterone exerts a variety of actions in the central nervous system under physiological and pathological conditions. As in other tissues, progesterone acts in the brain through classical progesterone receptors and through alternative mechanisms. Here, we review the role of progesterone as a regulator of kinases and phosphatases, such as extracellular-signal regulated kinases, phosphoinositide 3-kinase, Akt, glycogen synthase kinase 3, protein phosphatase 2A and phosphatase and tensin homolog deleted on chromosome 10. In addition, we analyzed the effects of progesterone on the phosphorylation of Tau, a protein that is involved in microtubule stabilization in neurons.
Collapse
|
25
|
Interactions of estradiol and insulin-like growth factor-I signalling in the nervous system: new advances. PROGRESS IN BRAIN RESEARCH 2010; 181:251-72. [PMID: 20478442 DOI: 10.1016/s0079-6123(08)81014-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Estradiol and insulin-like growth factor-I (IGF-I) interact in the brain to regulate a variety of developmental and neuroplastic events. Some of these interactions are involved in the control of hormonal homeostasis and reproduction. However, the interactions may also potentially impact on affection and cognition by the regulation of adult neurogenesis in the hippocampus and by promoting neuroprotection under neurodegenerative conditions. Recent studies suggest that the interaction of estradiol and IGF-I is also relevant for the control of cholesterol homeostasis in neural cells. The molecular mechanisms involved in the interaction of estradiol and IGF-I include the cross-regulation of the expression of estrogen and IGF-I receptors, the regulation of estrogen receptor-mediated transcription by IGF-I and the regulation of IGF-I receptor signalling by estradiol. Current investigations are evidencing the role exerted by key signalling molecules, such as glycogen synthase kinase 3 and beta-catenin, in the cross-talk of estrogen receptors and IGF-I receptors in neural cells.
Collapse
|
26
|
Hiney JK, Srivastava VK, Les Dees W. Insulin-like growth factor-1 stimulation of hypothalamic KiSS-1 gene expression is mediated by Akt: effect of alcohol. Neuroscience 2009; 166:625-32. [PMID: 20034543 DOI: 10.1016/j.neuroscience.2009.12.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Revised: 12/01/2009] [Accepted: 12/10/2009] [Indexed: 02/06/2023]
Abstract
Kisspeptin, as well as insulin-like growth factor-1 (IGF-1), act centrally to stimulate luteinizing hormone-releasing hormone (LHRH) secretion at puberty. IGF-1 can induce KiSS-1 gene expression as an early pubertal event; however, the signaling pathway mediating this effect is not known. Since alcohol (ALC) blocks IGF-1 induced LHRH release acutely, we assessed whether this drug could affect IGF-1 stimulated prepubertal KiSS-1 gene expression following a binge type of exposure. Immature female rats were administered either ALC (3 g/kg) or water via gastric gavage at 07.30 h. At 09.00 h the ALC and control groups were subdivided where half received either saline or IGF-1 (200 ng) into the third ventricle. A second dose of ALC (1.5, 2 and 3 g/kg) or water was administered at 11.30 h. These regimens produced moderate blood alcohol concentrations of 77, 89 and 117 mg/dl, respectively, over the time course of the experiment. Rats were sacrificed 6 h after the IGF-1 injection and tissues containing the anteroventral periventricular (AVPV) and arcuate (ARC) nuclei were collected. IGF-1 stimulated (P<0.01) KiSS-1 gene expression in the AVPV nucleus at 6 h, but did not affect expression of the kisspeptin receptor, GPR54. While ALC did not alter basal expression of either gene, its dose dependently blocked IGF-1-induced KiSS-1 gene expression in the AVPV nucleus. No changes were observed in the ARC nucleus. Assessment of IGF-1 signaling indicated that the acute administration of IGF-1, ALC, or both did not alter the basal expression of IGF-1 receptor protein. However, IGF-1 stimulated (P<0.05) phosphorylated Akt protein over basal levels, an action blocked by ALC. Our results indicate that the IGF-1 induction of KiSS-1 gene expression is mediated by Akt activation, and that ALC alters this important prepubertal action of IGF-1.
Collapse
Affiliation(s)
- J K Hiney
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A and M University College Station, TX 77843-4458, USA.
| | | | | |
Collapse
|
27
|
Kelly MJ, Rønnekleiv OK. Control of CNS neuronal excitability by estrogens via membrane-initiated signaling. Mol Cell Endocrinol 2009; 308:17-25. [PMID: 19549588 PMCID: PMC2701913 DOI: 10.1016/j.mce.2009.03.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 02/24/2009] [Accepted: 03/05/2009] [Indexed: 10/24/2022]
Abstract
It is well known that many of the actions of 17beta-estradiol (E2) in the central nervous system (CNS) are mediated via intracellular receptor/transcription factors that interact with steroid response elements on target genes. However, there is compelling evidence for membrane-associated steroid receptors for E2 in hypothalamic and other brain neurons. Indeed, we are just beginning to understand how E2 signals via membrane receptors, and how these signals impact not only membrane excitability but also gene transcription in neurons. We know that E2 can rapidly alter neuronal activity within seconds, indicating that some cellular effects can occur via membrane-delimited events. In addition, E2 can affect second messenger systems including calcium mobilization and a plethora of kinases to alter cell signaling. This review will concentrate on rapid membrane-initiated and intracellular signaling by E2 in the hypothalamus and hippocampus, the nature of receptors involved and how they contribute to CNS functions.
Collapse
Affiliation(s)
- Martin J. Kelly
- Department of Physiology and Pharmacology, L334, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA. E-mail, ; fax 503-494-4352, phone 503-494-5833
| | - Oline K. Rønnekleiv
- Department of Physiology and Pharmacology, L334, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA. E-mail, ; fax 503-494-4352, phone 503-494-5833
- Department of Anesthesiology and Perioperative Medicine, L334, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA. E-mail, ; fax 503-494-4352, phone 503-494-5840
- Division of Neuroscience, Oregon Regional Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| |
Collapse
|
28
|
Acs P, Kipp M, Norkute A, Johann S, Clarner T, Braun A, Berente Z, Komoly S, Beyer C. 17beta-estradiol and progesterone prevent cuprizone provoked demyelination of corpus callosum in male mice. Glia 2009; 57:807-14. [PMID: 19031445 DOI: 10.1002/glia.20806] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sex hormones, for example, estrogen and progesterone, are thought to affect and delay progression of multiple sclerosis (MS) in pregnant women. Although both steroid hormones are neuroprotective in the brain and elevated during pregnancy, only estrogen was tested in clinical trials. To evaluate the role of 17beta-estradiol (E) and progesterone (P) in prevention demyelination, young adult male mice were fed with cuprizone for a defined time interval and simultaneously treated with steroids by repeated injections into the neck region. The status of myelination was analyzed by magnetic resonance imaging and conventional histological staining. The individual application of E and P resulted only in a moderate prevention of demyelination in the corpus callosum (CC). The combined treatment with both steroid hormones counteracted the process of demyelination. Expression of the mature (PLP and MBP) and premature (PDGF-alpha-R) oligodendrocyte markers were significantly increased after hormone application in the affected CC. In addition, both hormones stimulated astrogliosis and the expression of IGF-1. Microglial invasion in demyelinated CC was pronounced and additionally localized in the midline of CC after hormone treatment. These data show that sex steroids can protect the brain from demyelination and stimulate remyelination. It appears that only the administration of both hormones is fully effective. The beneficial steroid effect requires interactions with oligodendrocytes possibly by preventing their degeneration or recruitment from precursor cells which are stimulated to remyelinated fibers. The positive hormonal influence on myelination in the CNS may be a future therapeutically strategy for the treatment of MS.
Collapse
Affiliation(s)
- Peter Acs
- Department of Neurology, Faculty of Medicine, University of Pécs, Rét u. 2, Pécs, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Srivastava VK, Hiney JK, Dees WL. Short-term alcohol administration alters KiSS-1 gene expression in the reproductive hypothalamus of prepubertal female rats. Alcohol Clin Exp Res 2009; 33:1605-14. [PMID: 19519717 DOI: 10.1111/j.1530-0277.2009.00992.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Kisspeptins bind to the G-protein-coupled receptor (GPR54) to activate hypothalamic luteinizing hormone releasing hormone (LHRH) secretion at the time of puberty. Alcohol (ALC) causes depressed prepubertal LHRH release, resulting in depressed luteinizing hormone (LH) secretion and delayed puberty. Because KiSS-1 and GPR54 are important to the onset of puberty, we assessed the effects of chronic ALC administration on basal expression of these puberty-related genes within the reproductive hypothalamus, as well as hormones and transduction signaling pathways contributing to their activity. METHODS Immature female rats were fed a liquid diet containing ALC for 6 days beginning when 27 days old. Controls received either companion isocaloric liquid diet or rat chow and water. Animals were decapitated on day 33, in the late juvenile stage of development. Blood was collected for the assessment of serum hormone levels. Brain tissues containing the anteroventral periventricular (AVPV) and arcuate (ARC) nuclei were obtained for assessing expression of specific puberty-related genes and proteins. RESULTS KiSS-1 mRNA levels in the AVPV and ARC nuclei were suppressed (p < 0.001) in the ALC-treated rats. GPR54 gene and protein expressions were both modestly increased (p < 0.05) in AVPV nucleus, but not in ARC nucleus. Alcohol exposure also resulted in suppressed serum levels of insulin-like growth factor-1 (IGF-1), LH, and estradiol (E(2)). As IGF-1, in the presence of E(2), can induce expression of the KiSS-1 gene, we assessed the potential for ALC to alter IGF-1 signaling in the reproductive hypothalamus. IGF-1 receptor gene and protein expressions were not altered. However, protein expression of phosphorylated Akt, a transduction signal used by IGF-1, was suppressed in the AVPV (p < 0.05) and ARC (p < 0.01) nuclei. CONCLUSIONS Alcohol causes suppressed KiSS-1 gene expression in the reproductive hypothalamus; hence, contributing to this drug's ability to cause suppressed LHRH secretion and disruption of the pubertal process. We suggest that this action, at least in part, is through altered IGF-1 signaling.
Collapse
Affiliation(s)
- Vinod K Srivastava
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | | | | |
Collapse
|
30
|
Marin R, Díaz M, Alonso R, Sanz A, Arévalo MA, Garcia-Segura LM. Role of estrogen receptor alpha in membrane-initiated signaling in neural cells: interaction with IGF-1 receptor. J Steroid Biochem Mol Biol 2009; 114:2-7. [PMID: 19167493 DOI: 10.1016/j.jsbmb.2008.12.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 12/31/2008] [Indexed: 12/25/2022]
Abstract
The mechanisms of action of estradiol in the nervous system involve nuclear-initiated steroid signaling and membrane-initiated steroid signaling. Estrogen receptors (ERs) are involved in both mechanisms. ERalpha interacts with the signaling of IGF-1 receptor in neural cells: ERalpha transcriptional activity is regulated by IGF-1 receptor signaling and estradiol regulates IGF-1 receptor signaling. The interaction between ERalpha and the IGF-1 receptor in the brain may occur at the plasma membrane of neurons and glial cells. Caveolin-1 may provide the scaffolding for the interaction of different membrane-associated molecules, including voltage-dependent anion channel, ERalpha and IGF-I receptor.
Collapse
Affiliation(s)
- Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Physiology & Institute of Biomedical Technologies, University of La Laguna, School of Medicine, Santa Cruz de Tenerife, Spain
| | | | | | | | | | | |
Collapse
|
31
|
Guerra-Araiza C, Amorim MA, Pinto-Almazán R, González-Arenas A, Campos MG, Garcia-Segura LM. Regulation of the phosphoinositide-3 kinase and mitogen-activated protein kinase signaling pathways by progesterone and its reduced metabolites in the rat brain. J Neurosci Res 2009; 87:470-81. [DOI: 10.1002/jnr.21848] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
32
|
González-Flores O, Etgen AM, Komisaruk BK, Gómora-Arrati P, Macias-Jimenez A, Lima-Hernández FJ, Garcia-Juárez M, Beyer C. Antagonists of the protein kinase A and mitogen-activated protein kinase systems and of the progestin receptor block the ability of vaginocervical/flank-perineal stimulation to induce female rat sexual behaviour. J Neuroendocrinol 2008; 20:1361-7. [PMID: 19094083 DOI: 10.1111/j.1365-2826.2008.01794.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Brief vaginocervical stimulation using a glass rod (VCS) combined with manual flank-perineal stimulation (FS) rapidly (within 5 min) induced both receptive and proceptive behavioural responses to males in ovariectomised, oestrogen-primed rats. This receptive-proceptive response to males, resulting from a single brief (5-s duration) instance of manual VCS + FS, declined markedly within 4 h. However, the decline was prevented if the females were mounted by males immediately after the manual VCS + FS and 2 h later. We tested the participation of the cAMP-dependent protein kinase A system and the mitogen-activated protein kinase (MAPK) system in the response to VCS + FS by infusing either 100 ng of Rp-adenosine 3',5'-cyclic monophosphorothiate triethylamonium salt (a protein kinase A blocker) or 3.3 microg of PD98059 (a MAPK blocker) i.c.v. 15 min prior to VCS + FS. Both inhibitors blocked the ability of VCS + FS to induce the proceptive-receptive responses to males at all testing intervals. In experiment 2, systemic administration of 5 mg of RU486 1 h before VCS + FS also blocked the ability of VCS + FS to induce the proceptive-receptive responses to males. The present findings suggest that both VCS + FS and mating stimuli provided by males release neurotransmitters and neuromodulators that trigger the protein kinase A and the MAPK signalling systems, which interact with the progestin receptor to rapidly (within 5 min) induce proceptive-receptive behaviour in females.
Collapse
Affiliation(s)
- O González-Flores
- Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Mo B, Callegari E, Telefont M, Renner KJ. Estrogen regulation of proteins in the rat ventromedial nucleus of the hypothalamus. J Proteome Res 2008; 7:5040-8. [PMID: 18841879 DOI: 10.1021/pr8005974] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The effects of estradiol (E2) on the expression of proteins in the pars lateralis of the ventromedial nucleus of the hypothalamus (VMNpl) in ovariectomized rats was studied using 2-dimensional gel electrophoresis followed by RPLC-nanoESI-MS/MS. E2 treatment resulted in the up-regulation of 29 identified proteins. Many of these proteins are implicated in the promotion of neuronal plasticity and signaling.
Collapse
Affiliation(s)
- Bing Mo
- Department of Biology and Neuroscience Group, University of South Dakota, Vermillion, South Daklota 57069, USA
| | | | | | | |
Collapse
|
34
|
Sanz A, Carrero P, Pernía O, Garcia-Segura LM. Pubertal maturation modifies the regulation of insulin-like growth factor-I receptor signaling by estradiol in the rat prefrontal cortex. Dev Neurobiol 2008; 68:1018-28. [PMID: 18446778 DOI: 10.1002/dneu.20641] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The transition from adolescence to adulthood is accompanied by substantial plastic modifications in the cerebral cortex, including changes in the growth and retraction of neuronal processes and in the rate of synaptic formation and neuronal loss. Some of these plastic changes are prevented in female rats by prepubertal ovariectomy. The ovarian hormone estradiol modulates neuronal differentiation and survival and these effects are in part mediated by the interaction with insulin-like growth factor-I (IGF-I). In this study, we have explored whether the activation by estradiol of some components of IGF-I receptor signaling is altered in the prefrontal cortex during puberty. Estradiol administration to rats ovariectomized after puberty resulted, 24 h after the hormonal administration, in a sustained phosphorylation of Akt and glycogen synthase kinase 3 beta in the prefrontal cortex. However, this hormonal effect was not observed in animals ovariectomized before puberty. These findings suggest that during pubertal maturation there is a programming by ovarian hormones of the future regulatory actions of estradiol on IGF-I receptor signaling in the prefrontal cortex. The modification in the regulation of IGF-I receptor signaling by estradiol during pubertal maturation may have implications for the developmental changes occurring in the prefrontal cortex in the transition from adolescence to adulthood.
Collapse
Affiliation(s)
- Amaya Sanz
- Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC, E-28002 Madrid, Spain
| | | | | | | |
Collapse
|
35
|
Garcia-Segura LM, Lorenz B, DonCarlos LL. The role of glia in the hypothalamus: implications for gonadal steroid feedback and reproductive neuroendocrine output. Reproduction 2008; 135:419-29. [PMID: 18367504 DOI: 10.1530/rep-07-0540] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Neuron-to-glia, glia-to-neuron, and glia-to-glia communication are implicated in the modulation of neuronal activity and synaptic transmission relevant to reproduction. Glial cells play an important role in neuroendocrine regulation and participate in the sexual differentiation of neuronal connectivity of brain regions involved in the control of reproductive neuroendocrine output. During puberty, modifications in the morphology and chemistry of astrocytes and tanycytes in the hypothalamus and median eminence influence the maturation of the neuronal circuits controlling the secretion of GnRH. During adult reproductive life, the glial cells participate in the transient remodeling of neuronal connectivity in the preoptic area, the arcuate nucleus, the median eminence, and other brain regions involved in the control of reproduction. Gonadal hormones regulate glial plasticity by direct and indirect effects and regulate various other endocrine signals, local soluble factors and adhesion molecules that also affect glial function and glia-to-neuron communication. The glial cells, therefore, are central to the coordination of endocrine and local inputs that bring about neural plasticity and adapt reproductive capacity to homeostatic signals.
Collapse
|
36
|
Ojeda SR, Lomniczi A, Sandau US. Glial-gonadotrophin hormone (GnRH) neurone interactions in the median eminence and the control of GnRH secretion. J Neuroendocrinol 2008; 20:732-42. [PMID: 18601696 DOI: 10.1111/j.1365-2826.2008.01712.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A wealth of information now exists showing that glial cells are actively involved in the cell-cell communication process generating and disseminating information within the central nervous system. In the hypothalamus, two types of glial cells, astrocytes and ependymal cells lining the latero-ventral portion of the third ventricle (known as tanycytes), regulate the secretory activity of neuroendocrine neurones. This function, initially described for astrocytes apposing magnocellular neurones, has been more recently characterised for neurones secreting gonadotrophin hormone-releasing hormone (GnRH). The available evidence suggests that glial cells of the median eminence regulate GnRH secretion via two related mechanisms. One involves the production of growth factors acting via receptors with tyrosine kinase activity. The other involves plastic rearrangements of glia-GnRH neurone adhesiveness. GnRH axons reach the median eminence, at least in part, directed by basic fibroblast growth factor. Their secretory activity is facilitated by insulin-like growth factor 1 and members of the epidermal growth factor family. A structural complement to these soluble molecules is provided by at least three cell-cell adhesion systems endowed with signalling capabilities. One of them uses the neuronal cell adhesion molecule (NCAM), another employs the synaptic cell adhesion molecule (SynCAM), and the third one consists of neuronal contactin interacting with glial receptor-like protein tyrosine phosphatase-beta. It is envisioned that, within the median eminence, soluble factors and adhesion molecules work coordinately to control delivery of GnRH to the portal vasculature.
Collapse
Affiliation(s)
- S R Ojeda
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | | | | |
Collapse
|
37
|
Gore AC. Developmental programming and endocrine disruptor effects on reproductive neuroendocrine systems. Front Neuroendocrinol 2008; 29:358-74. [PMID: 18394690 PMCID: PMC2702520 DOI: 10.1016/j.yfrne.2008.02.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2007] [Revised: 02/21/2008] [Accepted: 02/22/2008] [Indexed: 01/01/2023]
Abstract
The ability of a species to reproduce successfully requires the careful orchestration of developmental processes during critical time points, particularly the late embryonic and early postnatal periods. This article begins with a brief presentation of the evidence for how gonadal steroid hormones exert these imprinting effects upon the morphology of sexually differentiated hypothalamic brain regions, the mechanisms underlying these effects, and their implications in adulthood. Then, I review the evidence that aberrant exposure to hormonally-active substances such as exogenous endocrine-disrupting chemicals (EDCs), may result in improper hypothalamic programming, thereby decreasing reproductive success in adulthood. The field of endocrine disruption has shed new light on the discipline of basic reproductive neuroendocrinology through studies on how early life exposures to EDCs may alter gene expression via non-genomic, epigenetic mechanisms, including DNA methylation and histone acetylation. Importantly, these effects may be transmitted to future generations if the germline is affected via transgenerational, epigenetic actions. By understanding the mechanisms by which natural hormones and xenobiotics affect reproductive neuroendocrine systems, we will gain a better understanding of normal developmental processes, as well as develop the potential ability to intervene when development is disrupted.
Collapse
Affiliation(s)
- Andrea C Gore
- Division of Pharmacology and Toxicology, Institute for Neuroscience and Institute for Cellular and Molecular Biology, The University of Texas at Austin, 1 University Station A1915, Austin, TX 78712, USA.
| |
Collapse
|
38
|
Bibliography. Current world literature. Growth and development. Curr Opin Endocrinol Diabetes Obes 2008; 15:79-101. [PMID: 18185067 DOI: 10.1097/med.0b013e3282f4f084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Abstract
This paper is the 29th consecutive installment of the annual review of research concerning the endogenous opioid system, now spanning 30 years of research. It summarizes papers published during 2006 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurological disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY 11367, United States.
| |
Collapse
|
40
|
Gómora-Arrati P, Beyer C, Lima-Hernández FJ, Gracia ME, Etgen AM, González-Flores O. GnRH mediates estrous behavior induced by ring A reduced progestins and vaginocervical stimulation. Behav Brain Res 2007; 187:1-8. [PMID: 17888527 DOI: 10.1016/j.bbr.2007.08.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 06/09/2007] [Accepted: 08/20/2007] [Indexed: 10/22/2022]
Abstract
The present study was designed to assess the participation of gonadotropin-releasing hormone (GnRH) in the display of estrous behavior induced by application of vaginal-cervical stimulation (VCS) and by the intracerebroventricular (icv) administration of progesterone and its ring A-reduced metabolites to ovariectomized (ovx), estradiol benzoate (E2B) primed rats. Icv injection of Antide, a GnRH-1 receptor antagonist, significantly depressed lordosis behavior in ovx, E2B-primed rats treated with icv GnRH. Application of VCS to ovx, E2B-primed rats facilitated both lordosis and proceptivity. These behavioral responses were significantly depressed by the icv administration of Antide. Similarly, icv Antide blocked the stimulatory effect on both lordosis and proceptive behaviors elicited by progesterone and its ring A-reduced metabolites: 5alpha-pregnandione (5alpha-DHP), 5alpha-pregnan-3alpha-ol-20-one (5alpha,3alpha-Pgl) and 5beta-pregnan-3beta-hydroxy-20-one (5beta,3beta-Pgl) in ovx, E2B-primed rats. By contrast, icv injection of Antide failed to interfere with the facilitatory effect of the synthetic progestin megestrol acetate on lordosis and proceptive behaviors. This progestin is not reduced in ring A. The results suggest that GnRH release is an important process in the chain of events leading to the display of estrous behavior in response to progesterone, its ring A-reduced metabolites, and VCS in female rats.
Collapse
Affiliation(s)
- Porfirio Gómora-Arrati
- Centro de Investigación en Reproducción Animal, CINVESTAV Universidad Autónoma de Tlaxcala. Apdo. 62, Tlaxcala, c.p. 90000, Mexico
| | | | | | | | | | | |
Collapse
|
41
|
Mendez P, Wandosell F, Garcia-Segura LM. Cross-talk between estrogen receptors and insulin-like growth factor-I receptor in the brain: cellular and molecular mechanisms. Front Neuroendocrinol 2006; 27:391-403. [PMID: 17049974 DOI: 10.1016/j.yfrne.2006.09.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Revised: 08/11/2006] [Accepted: 09/01/2006] [Indexed: 01/02/2023]
Abstract
Accumulating evidence suggests that insulin-like growth factor-I (IGF-I) and estradiol interact to regulate neural function. In this review, we focus on the cellular and molecular mechanisms involved in this interaction. The expression of estrogen receptors (ERs) and IGF-I receptor is cross-regulated in the central nervous system and many neurons and astrocytes coexpress both receptors. Furthermore, estradiol activates IGF-I receptor and its intracellular signaling. This effect may involve classical ERs since recent findings suggest that ERalpha may affect IGF-I actions in the brain by a direct interaction with some of the components of IGF-I signaling. In turn, IGF-I may regulate ER transcriptional activity in neuronal cells. In conclusion, ERs appear to be part of the signaling mechanism of IGF-I, and IGF-I receptor part of the mechanism of estradiol signaling in the nervous system.
Collapse
Affiliation(s)
- Pablo Mendez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), E-28002 Madrid, Spain
| | | | | |
Collapse
|