1
|
Song Y, Zhang K, Zhang J, Li Q, Huang N, Ma Y, Hou N, Han F, Kan C, Sun X. Epigenetic regulation of nuclear receptors: Implications for endocrine-related diseases and therapeutic strategies. Genes Dis 2025; 12:101481. [PMID: 40290121 PMCID: PMC12022648 DOI: 10.1016/j.gendis.2024.101481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/20/2024] [Accepted: 11/03/2024] [Indexed: 04/30/2025] Open
Abstract
The expression and function of the receptor are controlled by epigenetic changes, such as DNA methylation, histone modification, and noncoding RNAs. These modifications play a pivotal role in receptor activity and can lead to or exacerbate endocrine-related diseases. This review examines the epigenetic alterations of nuclear receptors and their significant impact on conditions such as diabetes, thyroid disorders, and endocrine-related tumors. It highlights current therapies targeting these epigenetic mechanisms, including gene editing, epigenetic drugs, and various other therapeutic approaches. This review offers fresh insight into the mechanisms of endocrine-associated disorders, highlighting the latest progress in the development of novel epigenetic therapies that can be used to address receptor-endocrine interactions.
Collapse
Affiliation(s)
- Yixin Song
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Qinying Li
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Na Huang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Yujie Ma
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| |
Collapse
|
2
|
Chrisp R, Masterson M, Pope R, Roberts CJ, Collins HM, Watson DJG, O'Neil D, Aagaard KM, Gibson CL, Heery DM, Moran PM. Sex-specific attenuation of constant light-induced memory impairment and Clock gene expression in brain in hepatic Npas2 knockout mice. Sci Rep 2025; 15:8347. [PMID: 40069567 PMCID: PMC11897300 DOI: 10.1038/s41598-025-92938-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
NPAS2 (Neuronal PAS Domain Protein 2) is a component of the core circadian clock and the coordinated activity between central brain and peripheral liver clock proteins postulated to be instrumental for linking behaviour and metabolism. We investigated a conditional liver-specific knockout mouse model (Npas2-/- or cKO) to explore its function in activity, circadian rhythms and cognition (novel object recognition-NOR). Circadian rhythms showed no genotype differences. Constant-light reduced NOR in floxxed controls but remarkably not in Npas2-/- mice, particularly females. Consistent with entrainment of systemic and central circadian biology, Npas2-/- mice showed altered expression of circadian gene Clock in frontal cortex. Sex differences independent of genotype were found in expression of circadian genes Clock, Bmal1 and Reverb-b in brain. Sex differences in Clock were absent in Npas2-/- mice. Females showed greater period length and phase response to constant light independently of genotype. The data suggest that a role for peripheral NPAS2 in constant light-induced memory impairment in females, and potential mediation by altered cortical circadian Clock gene expression, merit further investigation. These findings have implications for the interaction between peripheral and central circadian clocks, circadian sex differences and the deleterious effects of constant light on cognition.
Collapse
Affiliation(s)
- Ruby Chrisp
- Gene Regulation and RNA Biology Laboratory, School of Pharmacy, BioDiscovery Institute, University Park, University of Nottingham, Nottingham, NG7 2RD, UK
- School of Psychology, University Park, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Mitchell Masterson
- Gene Regulation and RNA Biology Laboratory, School of Pharmacy, BioDiscovery Institute, University Park, University of Nottingham, Nottingham, NG7 2RD, UK
- School of Psychology, University Park, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Rebecca Pope
- School of Psychology, University Park, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Christopher J Roberts
- Gene Regulation and RNA Biology Laboratory, School of Pharmacy, BioDiscovery Institute, University Park, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Hilary M Collins
- Gene Regulation and RNA Biology Laboratory, School of Pharmacy, BioDiscovery Institute, University Park, University of Nottingham, Nottingham, NG7 2RD, UK
| | - David J G Watson
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Derek O'Neil
- Division of Maternal-Fetal Medicine, Departments of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Bioinformatics Research Laboratory, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Departments of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Bioinformatics Research Laboratory, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Claire L Gibson
- School of Psychology, University Park, University of Nottingham, Nottingham, NG7 2RD, UK
| | - David M Heery
- Gene Regulation and RNA Biology Laboratory, School of Pharmacy, BioDiscovery Institute, University Park, University of Nottingham, Nottingham, NG7 2RD, UK.
| | - Paula M Moran
- School of Psychology, University Park, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
3
|
Kwag E, Park SJ, Lee JH, Lee JY, Khang R, Shin JH. Upregulation of p52-ZER6 (ZNF398) increases reactive oxygen species by suppressing metallothionein-3 in neuronal cells. Biochem Biophys Res Commun 2025; 748:151316. [PMID: 39809138 DOI: 10.1016/j.bbrc.2025.151316] [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: 11/04/2024] [Revised: 12/27/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
ZNF398/ZER6 belongs to the Krüppel-associated box (KRAB) domain-containing zinc finger proteins (K-ZNFs), the largest family of transcriptional repressors in higher organisms. ZER6 exists in two isoforms, p52 and p71, generated through alternative splicing. Our investigation revealed that p71-ZER6 is abundantly expressed in the stomach, kidney, liver, heart, and brown adipose tissue, while p52-ZER6 is predominantly found in the stomach and brain. The role of p52-ZER6 in neurons has remained unclear. Leveraging open-source RNA-seq data, we identified metallothionein 3 (MT3) as a target gene of p52-ZER6 in mouse hippocampal neuronal HT-22 cells. Through chromatin immunoprecipitation assays, we identified the putative DNA-binding motif (CTAGGGGGGTTGTTATCTCTTTGG) of p52-ZER6 in the promoter region of MT3. Furthermore, we demonstrated an interaction between p52-ZER6 and estrogen receptor alpha (ERα) in the nucleus of SH-SY5Y cells, which led to the inhibition of p52-ZER6's DNA occupancy on the promoter of the MT3 gene. MT3 is a cysteine-rich, low molecular-weight protein known for reducing oxidative stress, reactive oxygen species (ROS), and metal toxicity. Our study revealed that overexpression of p52-ZER6 reduced the levels of MT3, increasing ROS levels, which was mitigated by co-overexpression of ERα. Notably, we also observed upregulation of p52-ZER6 and reduction of MT3 in the cortex of 5xFAD, an Alzheimer's disease (AD) mouse model. These findings suggest a potential pathological mechanism involving p52-ZER6-mediated ROS production in AD pathogenesis.
Collapse
Affiliation(s)
- Eunsang Kwag
- Department of Pharmacology, Republic of Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 440-746, Republic of Korea
| | - Soo Jeong Park
- Department of Pharmacology, Republic of Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 440-746, Republic of Korea
| | - Jee-Ho Lee
- Department of Pharmacology, Republic of Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 440-746, Republic of Korea
| | - Ji-Yeong Lee
- Department of Pharmacology, Republic of Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 440-746, Republic of Korea
| | - Rin Khang
- Department of Pharmacology, Republic of Korea
| | - Joo-Ho Shin
- Department of Pharmacology, Republic of Korea; Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 440-746, Republic of Korea; Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, Republic of Korea.
| |
Collapse
|
4
|
Mitchnick KA, Nicholson K, Wideman C, Jardine K, Jamieson-Williams R, Creighton SD, Lacoursiere A, Milite C, Castellano S, Sbardella G, MacLusky NJ, Choleris E, Winters BD. The Lysine Acetyltransferase PCAF Functionally Interacts with Estrogen Receptor Alpha in the Hippocampus of Gonadally Intact Male-But Not Female-Rats to Enhance Short-Term Memory. J Neurosci 2024; 44:e1574232024. [PMID: 39138001 PMCID: PMC11376336 DOI: 10.1523/jneurosci.1574-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 07/05/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Acetylation of histone proteins by histone acetyltransferases (HATs), and the resultant change in gene expression, is a well-established mechanism necessary for long-term memory (LTM) consolidation, which is not required for short-term memory (STM). However, we previously demonstrated that the HAT p300/CBP-associated factor (PCAF) also influences hippocampus (HPC)-dependent STM in male rats. In addition to their epigenetic activity, HATs acetylate nonhistone proteins involved in nongenomic cellular processes, such as estrogen receptors (ERs). Given that ERs have rapid, nongenomic effects on HPC-dependent STM, we investigated the potential interaction between ERs and PCAF for STM mediated by the dorsal hippocampus (dHPC). Using a series of pharmacological agents administered directly into the dHPC, we reveal a functional interaction between PCAF and ERα in the facilitation of short-term object-in-place memory in male but not female rats. This interaction was specific to ERα, while ERβ agonism did not enhance STM. It was further specific to dHPC STM, as the effect was not present in the dHPC for LTM or in the perirhinal cortex. Further, while STM required local (i.e., dHPC) estrogen synthesis, the facilitatory interaction effect appeared independent of estrogens. Finally, western blot analyses demonstrated that PCAF activation in the dHPC rapidly (5 min) activated downstream estrogen-related cell signaling kinases (c-Jun N-terminal kinase and extracellular signal-related kinase). Collectively, these findings indicate that PCAF, which is typically implicated in LTM through epigenetic processes, also influences STM in the dHPC, possibly via nongenomic ER activity. Critically, this novel PCAF-ER interaction might exist as a male-specific mechanism supporting STM.
Collapse
Affiliation(s)
- Krista A Mitchnick
- Department of Psychology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Collaborative Neuroscience Program, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kate Nicholson
- Collaborative Neuroscience Program, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Cassidy Wideman
- Department of Psychology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Collaborative Neuroscience Program, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kristen Jardine
- Department of Psychology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Collaborative Neuroscience Program, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | | | - Samantha D Creighton
- Department of Psychology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Collaborative Neuroscience Program, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Allison Lacoursiere
- Department of Psychology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Ciro Milite
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno, Fisciano I-84084, Italy
| | - Sabrina Castellano
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno, Fisciano I-84084, Italy
| | - Gianluca Sbardella
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno, Fisciano I-84084, Italy
| | - Neil J MacLusky
- Collaborative Neuroscience Program, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Elena Choleris
- Department of Psychology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Collaborative Neuroscience Program, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Boyer D Winters
- Department of Psychology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
- Collaborative Neuroscience Program, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
5
|
Wang L, Xu S, Chen R, Ding Y, Liu M, Hou C, Wu Z, Men X, Bao M, He B, Li S. Exploring the causal association between epigenetic clocks and menopause age: insights from a bidirectional Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1429514. [PMID: 39247918 PMCID: PMC11377254 DOI: 10.3389/fendo.2024.1429514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/02/2024] [Indexed: 09/10/2024] Open
Abstract
Background Evidence suggests a connection between DNA methylation (DNAm) aging and reproductive aging. However, the causal relationship between DNAm and age at menopause remains uncertain. Methods Employing established DNAm epigenetic clocks, such as DNAm Hannum age acceleration (Hannum), Intrinsic epigenetic age acceleration (IEAA), DNAm-estimated granulocyte proportions (Gran), DNAm GrimAge acceleration (GrimAgeAccel), DNAm PhenoAge acceleration (PhenoAgeAccel), and DNAm-estimated plasminogen activator inhibitor-1 levels (DNAmPAIadjAge), a bidirectional Mendelian randomization (MR) study was carried out to explore the potential causality between DNAm and menopausal age. The primary analytical method used was the inverse variance weighted (IVW) estimation model, supplemented by various other estimation techniques. Results DNAm aging acceleration or deceleration, as indicated by Hannum, IEAA, Gran, GrimAgeAccel, PhenoAgeAccel, and DNAmPAIadjAge, did not exhibit a statistically significant causal effect on menopausal age according to forward MR analysis. However, there was a suggestive positive causal association between age at menopause and Gran (Beta = 0.0010; 95% confidence interval (CI): 0.0004, 0.0020) in reverse MR analysis. Conclusion The observed increase in granulocyte DNAm levels in relation to menopausal age could potentially serve as a valuable indicator for evaluating the physiological status at the onset of menopause.
Collapse
Affiliation(s)
- Ling Wang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Shuling Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Rumeng Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yining Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Menghua Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chunyan Hou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhu Wu
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Xiaoju Men
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Meihua Bao
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Binsheng He
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
McClarty BM, Rodriguez G, Dong H. Class 1 histone deacetylases differentially modulate memory and synaptic genes in a spatial and temporal manner in aged and APP/PS1 mice. Brain Res 2024; 1837:148951. [PMID: 38642789 PMCID: PMC11182336 DOI: 10.1016/j.brainres.2024.148951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Epigenetics plays a vital role in aging and Alzheimer's disease (AD); however, whether epigenetic alterations during aging can initiate AD and exacerbate AD progression remains unclear. In this study, using 3-, 12- and 18- month-old APP/PS1 mice and age matched WT littermates, we conducted a series of memory tests, measured synapse-related gene expression, class 1 histone deacetylases (HDACs) abundance, and H3K9ac levels at target gene promoters in the hippocampus and prefrontal cortex (PFC). Our results showed impaired recognition and long-term spatial memory in 18-month-old WT mice and impaired recognition, short-term working, and long-term spatial reference memory in 12-and 18- month-old APP/PS1 mice. These memory impairments are associated with changes of synapse-related gene (nr2a, glur1, glur2, psd95) expression, HDAC abundance, and H3K9ac levels; more specifically, increased HDAC2 was associated with synapse-related gene expression changes through modulation of H3K9ac at the gene promoters during aging and AD progression in the hippocampus. Conversely, increased HDAC3 was associated with synapse-related gene expression changes through modulation of H3K9ac at the gene promoters during AD progression in the PFC. These findings suggest memory impairments in aging and AD may associated with a differential HDAC modulation of synapse-related gene expression in the brain.
Collapse
Affiliation(s)
- Bryan M McClarty
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 7-103, Chicago, IL 60611, USA
| | - Guadalupe Rodriguez
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 7-103, Chicago, IL 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 7-103, Chicago, IL 60611, USA.
| |
Collapse
|
7
|
Feng Y, Qin J, Lu Y, Wang M, Wang S, Luo F. Suberoylanilide hydroxamic acid attenuates cognitive impairment in offspring caused by maternal surgery during mid-pregnancy. PLoS One 2024; 19:e0295096. [PMID: 38551911 PMCID: PMC10980197 DOI: 10.1371/journal.pone.0295096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/13/2024] [Indexed: 04/01/2024] Open
Abstract
Some pregnant women have to experience non-obstetric surgery during pregnancy under general anesthesia. Our previous studies showed that maternal exposure to sevoflurane, isoflurane, propofol, and ketamine causes cognitive deficits in offspring. Histone acetylation has been implicated in synaptic plasticity. Propofol is commonly used in non-obstetric procedures on pregnant women. Previous studies in our laboratory showed that maternal propofol exposure in pregnancy impairs learning and memory in offspring by disturbing histone acetylation. The present study aims to investigate whether HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) could attenuate learning and memory deficits in offspring caused by maternal surgery under propofol anesthesia during mid-pregnancy. Maternal rats were exposed to propofol or underwent abdominal surgery under propofol anesthesia during middle pregnancy. The learning and memory abilities of the offspring rats were assessed using the Morris water maze (MWM) test. The protein levels of histone deacetylase 2 (HDAC2), phosphorylated cAMP response-element binding (p-CREB), brain-derived neurotrophic factor (BDNF), and phosphorylated tyrosine kinase B (p-TrkB) in the hippocampus of the offspring rats were evaluated by immunofluorescence staining and western blot. Hippocampal neuroapoptosis was detected by TUNEL staining. Our results showed that maternal propofol exposure during middle pregnancy impaired the water-maze learning and memory of the offspring rats, increased the protein level of HDAC2 and reduced the protein levels of p-CREB, BDNF and p-TrkB in the hippocampus of the offspring, and such effects were exacerbated by surgery. SAHA alleviated the cognitive dysfunction and rescued the changes in the protein levels of p-CREB, BDNF and p-TrkB induced by maternal propofol exposure alone or maternal propofol exposure plus surgery. Therefore, SAHA could be a potential and promising agent for treating the learning and memory deficits in offspring caused by maternal nonobstetric surgery under propofol anesthesia.
Collapse
Affiliation(s)
- Yunlin Feng
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jia Qin
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanfei Lu
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Mengdie Wang
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shengqiang Wang
- Department of Anesthesiology, Yichun People’s Hospital, Yichun, China
| | - Foquan Luo
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Boyd HM, Frick KM, Kwapis JL. Connecting the Dots: Potential Interactions Between Sex Hormones and the Circadian System During Memory Consolidation. J Biol Rhythms 2023; 38:537-555. [PMID: 37464775 PMCID: PMC10615791 DOI: 10.1177/07487304231184761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Both the circadian clock and sex hormone signaling can strongly influence brain function, yet little is known about how these 2 powerful modulatory systems might interact during complex neural processes like memory consolidation. Individually, the molecular components and action of each of these systems have been fairly well-characterized, but there is a fundamental lack of information about how these systems cooperate. In the circadian system, clock genes function as timekeeping molecules that convey time-of-day information on a well-stereotyped cycle that is governed by the suprachiasmatic nucleus. Keeping time is particularly important to synchronize various physiological processes across the brain and body, including those that regulate memory consolidation. Similarly, sex hormones are powerful modulators of memory, with androgens, estrogens, and progestins, all influencing memory consolidation within memory-relevant brain regions like the hippocampus. Despite clear evidence that each system can influence memory individually, exactly how the circadian and hormonal systems might interact to impact memory consolidation remains unclear. Research investigating either sex hormone action or circadian gene function within memory-relevant brain regions has unveiled several notable places in which the two systems could interact to control memory. Here, we bring attention to known interactions between the circadian clock and sex hormone signaling. We then review sex hormone-mediated control of memory consolidation, highlighting potential nodes through which the circadian system might interact during memory formation. We suggest that the bidirectional relationship between these two systems is essential for proper control of memory formation based on an animal's hormonal and circadian state.
Collapse
Affiliation(s)
- Hannah M. Boyd
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| | - Karyn M. Frick
- Department of Psychology, University of Wisconsin–Milwaukee, Milwaukee, Wisconsin
| | - Janine L. Kwapis
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
9
|
Fabian CB, Seney ML, Joffe ME. Sex differences and hormonal regulation of metabotropic glutamate receptor synaptic plasticity. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 168:311-347. [PMID: 36868632 PMCID: PMC10392610 DOI: 10.1016/bs.irn.2022.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Striking sex differences exist in presentation and incidence of several psychiatric disorders. For example, major depressive disorder is more prevalent in women than men, and women who develop alcohol use disorder progress through drinking milestones more rapidly than men. With regards to psychiatric treatment responses, women respond more favorably to selective serotonin reuptake inhibitors than men, whereas men have better outcomes when prescribed tricyclic antidepressants. Despite such well-documented biases in incidence, presentation, and treatment response, sex as a biological variable has long been neglected in preclinical and clinical research. An emerging family of druggable targets for psychiatric diseases, metabotropic glutamate (mGlu) receptors are G-protein coupled receptors broadly distributed throughout the central nervous system. mGlu receptors confer diverse neuromodulatory actions of glutamate at the levels of synaptic plasticity, neuronal excitability, and gene transcription. In this chapter, we summarize the current preclinical and clinical evidence for sex differences in mGlu receptor function. We first highlight basal sex differences in mGlu receptor expression and function and proceed to describe how gonadal hormones, notably estradiol, regulate mGlu receptor signaling. We then describe sex-specific mechanisms by which mGlu receptors differentially modulate synaptic plasticity and behavior in basal states and models relevant for disease. Finally, we discuss human research findings and highlight areas in need of further research. Taken together, this review emphasizes how mGlu receptor function and expression can differ across sex. Gaining a more complete understanding of how sex differences in mGlu receptor function contribute to psychiatric diseases will be critical in the development of novel therapeutics that are effective in all individuals.
Collapse
Affiliation(s)
- Carly B Fabian
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - Marianne L Seney
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - Max E Joffe
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
10
|
Kaczynski P, van der Weijden V, Goryszewska-Szczurek E, Baryla M, Ulbrich SE, Waclawik A. Novel role for conceptus signals in mRNA expression regulation by DNA methylation in porcine endometrium during early pregnancy†. Biol Reprod 2022; 108:150-168. [PMID: 36322137 PMCID: PMC9843678 DOI: 10.1093/biolre/ioac193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/12/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
During early pregnancy, porcine conceptuses (the embryos with associated membranes) secrete estradiol-17β (E2)-their major signal for maternal recognition of pregnancy-and prostaglandin E2 (PGE2). Both hormones induce prominent changes of the endometrial transcriptome in vivo. Studies on endometrial pathologies have shown that E2 affects gene expression by epigenetic mechanisms related to DNA methylation. Herein, we determined the effects of E2 and PGE2 alone, and a combined E2 + PGE2 treatment administered into the uterine lumen in vivo on the expression and activity of DNA-methyltransferases (DNMTs) and on CpG methylation patterns of selected genes in porcine endometrium. To compare the effect of treatment with the physiological effect of pregnancy, endometria from day 12 pregnant/cyclic gilts were included. Both E2 and PGE2 significantly reduced the expression of DNMTs. Likewise, the expressions of DNMT1 and DNMT3A were decreased on day 12 of pregnancy compared to the estrous cycle. DNMT activity increased in endometrial samples following E2 treatment and in gilts on day 12 of pregnancy. Treatment with E2 alone and/or simultaneously with PGE2 altered endometrial DNA methylation of CpG sites of ADAMTS20, ADH1C, BGN, PSAT1, and WNT5A. Different CpG methylation patterns of ADAMTS20, BGN, DMBT1, RASSF1, and WNT5A were found in the endometrium on day 12 of pregnancy compared to day 12 of the estrous cycle. Significant correlations were detected between CpG methylation and gene expression for ADAMTS20, ADH1C, BGN, DMBT1, PSAT1, and WNT5A. Our results indicate that CpG methylation induced by embryonic signals may contribute to regulating endometrial gene expression during pregnancy establishment.
Collapse
Affiliation(s)
- Piotr Kaczynski
- Correspondence: Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland. Tel: +48895393111; E-mail: ; (A. Waclawik); Tel: +48895393180; E-mail: (P. Kaczynski)
| | - Vera van der Weijden
- ETH Zurich, Animal Physiology, Institute of Agricultural Sciences, Zurich, Switzerland
| | | | - Monika Baryla
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Susanne E Ulbrich
- ETH Zurich, Animal Physiology, Institute of Agricultural Sciences, Zurich, Switzerland
| | - Agnieszka Waclawik
- Correspondence: Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland. Tel: +48895393111; E-mail: ; (A. Waclawik); Tel: +48895393180; E-mail: (P. Kaczynski)
| |
Collapse
|
11
|
Hatakeyama D, Sunada H, Totani Y, Watanabe T, Felletár I, Fitchett A, Eravci M, Anagnostopoulou A, Miki R, Okada A, Abe N, Kuzuhara T, Kemenes I, Ito E, Kemenes G. Molecular and functional characterization of an evolutionarily conserved CREB-binding protein in the Lymnaea CNS. FASEB J 2022; 36:e22593. [PMID: 36251357 PMCID: PMC9828244 DOI: 10.1096/fj.202101225rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/30/2022] [Accepted: 09/26/2022] [Indexed: 01/12/2023]
Abstract
In eukaryotes, CREB-binding protein (CBP), a coactivator of CREB, functions both as a platform for recruiting other components of the transcriptional machinery and as a histone acetyltransferase (HAT) that alters chromatin structure. We previously showed that the transcriptional activity of cAMP-responsive element binding protein (CREB) plays a crucial role in neuronal plasticity in the pond snail Lymnaea stagnalis. However, there is no information on the molecular structure and HAT activity of CBP in the Lymnaea central nervous system (CNS), hindering an investigation of its postulated role in long-term memory (LTM). Here, we characterize the Lymnaea CBP (LymCBP) gene and identify a conserved domain of LymCBP as a functional HAT. Like CBPs of other species, LymCBP possesses functional domains, such as the KIX domain, which is essential for interaction with CREB and was shown to regulate LTM. In-situ hybridization showed that the staining patterns of LymCBP mRNA in CNS are very similar to those of Lymnaea CREB1. A particularly strong LymCBP mRNA signal was observed in the cerebral giant cell (CGC), an identified extrinsic modulatory interneuron of the feeding circuit, the key to both appetitive and aversive LTM for taste. Biochemical experiments using the recombinant protein of the LymCBP HAT domain showed that its enzymatic activity was blocked by classical HAT inhibitors. Preincubation of the CNS with such inhibitors blocked cAMP-induced synaptic facilitation between the CGC and an identified follower motoneuron of the feeding system. Taken together, our findings suggest a role for the HAT activity of LymCBP in synaptic plasticity in the feeding circuitry.
Collapse
Affiliation(s)
- Dai Hatakeyama
- Sussex NeuroscienceSchool of Life Sciences, University of SussexBrightonUK,Faculty of Pharmaceutical SciencesTokushima Bunri UniversityTokushimaJapan
| | - Hiroshi Sunada
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri UniversitySanukiJapan,Present address:
Advanced Medicine, Innovation and Clinical Research CentreTottori University HospitalYonagoJapan
| | - Yuki Totani
- Department of BiologyWaseda UniversityTokyoJapan
| | | | - Ildikó Felletár
- Sussex NeuroscienceSchool of Life Sciences, University of SussexBrightonUK
| | - Adam Fitchett
- Sussex NeuroscienceSchool of Life Sciences, University of SussexBrightonUK
| | - Murat Eravci
- Sussex NeuroscienceSchool of Life Sciences, University of SussexBrightonUK
| | - Aikaterini Anagnostopoulou
- Sussex NeuroscienceSchool of Life Sciences, University of SussexBrightonUK,Present address:
School of Life SciencesUniversity of WestminsterLondonUK
| | - Ryosuke Miki
- Faculty of Pharmaceutical SciencesTokushima Bunri UniversityTokushimaJapan
| | - Ayano Okada
- Faculty of Pharmaceutical SciencesTokushima Bunri UniversityTokushimaJapan
| | - Naoya Abe
- Faculty of Pharmaceutical SciencesTokushima Bunri UniversityTokushimaJapan
| | - Takashi Kuzuhara
- Faculty of Pharmaceutical SciencesTokushima Bunri UniversityTokushimaJapan
| | - Ildikó Kemenes
- Sussex NeuroscienceSchool of Life Sciences, University of SussexBrightonUK
| | - Etsuro Ito
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri UniversitySanukiJapan,Department of BiologyWaseda UniversityTokyoJapan
| | - György Kemenes
- Sussex NeuroscienceSchool of Life Sciences, University of SussexBrightonUK
| |
Collapse
|
12
|
Chromatin modifiers – Coordinators of estrogen action. Biomed Pharmacother 2022; 153:113548. [DOI: 10.1016/j.biopha.2022.113548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 11/20/2022] Open
|
13
|
Taxier LR, Philippi SM, York JM, LaDu MJ, Frick KM. The detrimental effects of APOE4 on risk for Alzheimer's disease may result from altered dendritic spine density, synaptic proteins, and estrogen receptor alpha. Neurobiol Aging 2022; 112:74-86. [PMID: 35051676 PMCID: PMC8976726 DOI: 10.1016/j.neurobiolaging.2021.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/08/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023]
Abstract
Women carriers of APOE4, the greatest genetic risk factor for late-onset Alzheimer's disease (AD), are at highest risk of developing AD, yet factors underlying interactions between APOE4 and sex are not well characterized. Here, we examined how sex and APOE3 or APOE4 genotypes modulate object and spatial memory, dendritic spine density and branching, and protein expression in 6-month-old male and female E3FAD and E4FAD mice (APOE+/+/5xFAD+/-). APOE4 negatively impacted object recognition and spatial memory, with male E3FADs exhibiting the best memory across 2 object-based tasks. In both sexes, APOE4 reduced basal dendritic spine density in the medial prefrontal cortex and dorsal hippocampus. APOE4 reduced dorsal hippocampal levels of PDS-95, synaptophysin, and phospho-CREB, yet increased levels of ERα. E4FAD females exhibited strikingly increased GFAP levels, in addition to the lowest levels of PSD-95 and pCREB. Overall, our results suggest that APOE4 negatively impacts object memory, dendritic spine density, and levels of hippocampal synaptic proteins and ERα. However, the general lack of sex differences or sex by genotype interactions suggests that the sex-specific effects of APOE4 on AD risk may be related to factors unexplored in the present study.
Collapse
Affiliation(s)
- Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee WI, USA
| | - Sarah M Philippi
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee WI, USA
| | - Jason M York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee WI, USA.
| |
Collapse
|
14
|
Mahajan V, Gujral P, Jain L, Ponnampalam AP. Differential Expression of Steroid Hormone Receptors and Ten Eleven Translocation Proteins in Endometrial Cancer Cells. Front Oncol 2022; 12:763464. [PMID: 35372016 PMCID: PMC8966408 DOI: 10.3389/fonc.2022.763464] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/16/2022] [Indexed: 11/26/2022] Open
Abstract
Steroid hormones govern the complex, cyclic changes of the endometrium, predominantly through their receptors. An interplay between steroid hormones and epigenetic mechanisms controls the dynamic endometrial gene regulation. Abnormalities in expression of genes and enzymes associated with steroid hormone signaling, contribute to a disturbed hormonal equilibrium. Limited evidence suggests the involvement of TET (Ten Eleven Translocation)-mediated DNA hydroxymethylation in endometrial cancer, with some data on the use of TET1 as a potential prognostic and diagnostic biomarker, however the mechanisms guiding it and its regulation remains unexplored. This study aims to explore the changes in the expressions of TETs and steroid hormone receptors in response to estrogen and progesterone in endometrial cancer cells. Gene expression was examined using real-time PCR and protein expression was quantified using fluorescent western blotting in endometrial cancer cell lines (AN3 and RL95-2). Results indicate that TET1 and TET3 gene and protein expression was cell-specific in cancer cell-lines. Protein expression of TET1 was downregulated in AN3 cells, while TET1 and TET3 expressions were both upregulated in RL95-2 cells in response to estrogen-progesterone. Further, a decreased AR expression in AN3 cells and an increased ERα and ERβ protein expressions in RL95-2 cells was seen in response to estrogen-progesterone. PR gene and protein expression was absent from both cancer cell-lines. Overall, results imply that expressions of steroid hormones, steroid-hormone receptors and TETs are co-regulated in endometrial cancer-cells. Further studies are needed to interpret how these mechanisms fit in with DNMTs and DNA methylation in regulating endometrial biology. Understanding the role of TETs and hydroxymethylation in steroid hormone receptor regulation is crucial to comprehend how these mechanisms work together in a broader context of epigenetics in the endometrium and its pathologies.
Collapse
Affiliation(s)
- Vishakha Mahajan
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Palak Gujral
- The Liggins Institute, University of Auckland, Auckland, New Zealand
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Lekha Jain
- The Liggins Institute, University of Auckland, Auckland, New Zealand
- Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Anna P. Ponnampalam
- The Liggins Institute, University of Auckland, Auckland, New Zealand
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- *Correspondence: Anna P. Ponnampalam,
| |
Collapse
|
15
|
Enriquez KD, Gupta AR, Hoffman EJ. Signaling Pathways and Sex Differential Processes in Autism Spectrum Disorder. Front Psychiatry 2021; 12:716673. [PMID: 34690830 PMCID: PMC8531220 DOI: 10.3389/fpsyt.2021.716673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/02/2021] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorders (ASDs) are a group of neurodevelopmental disorders associated with deficits in social communication and restrictive, repetitive patterns of behavior, that affect up to 1 in 54 children. ASDs clearly demonstrate a male bias, occurring ~4 times more frequently in males than females, though the basis for this male predominance is not well-understood. In recent years, ASD risk gene discovery has accelerated, with many whole-exome sequencing studies identifying genes that converge on common pathways, such as neuronal communication and regulation of gene expression. ASD genetics studies have suggested that there may be a "female protective effect," such that females may have a higher threshold for ASD risk, yet its etiology is not well-understood. Here, we review common biological pathways implicated by ASD genetics studies as well as recent analyses of sex differential processes in ASD using imaging genomics, transcriptomics, and animal models. Additionally, we discuss recent investigations of ASD risk genes that have suggested a potential role for estrogens as modulators of biological pathways in ASD, and highlight relevant molecular and cellular pathways downstream of estrogen signaling as potential avenues for further investigation.
Collapse
Affiliation(s)
- Kristen D Enriquez
- Program on Neurogenetics, Child Study Center, Yale University School of Medicine, New Haven, CT, United States
| | - Abha R Gupta
- Program on Neurogenetics, Child Study Center, Yale University School of Medicine, New Haven, CT, United States.,Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Ellen J Hoffman
- Program on Neurogenetics, Child Study Center, Yale University School of Medicine, New Haven, CT, United States.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
16
|
Di Zhang D, Wang YG, Liu CY, Wang ZH, Wang YF. Aminoguanidine ameliorates ovariectomy-induced neuronal deficits in rats by inhibiting AGE-mediated Aβ production. Acta Neurobiol Exp (Wars) 2021; 81:10-20. [PMID: 33949165 DOI: 10.21307/ane-2021-002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 10/08/2020] [Indexed: 11/11/2022]
Abstract
Advanced glycation end products (AGEs) have been reported to cause neurodegeneration, senile plaque formation and spatial learning and memory deficits. There is much evidence describing the beneficial effects of aminoguanidine (AG) on the central nervous system; AG is able to inhibit the receptor for AGEs and beta-amyloid (Aβ) deposition in the brain, thus preventing cognitive decline and neurodegeneration. In this study, we investigated whether AG protects against ovariectomy-induced neuronal deficits and Aβ deposition in rats. Animals in the ovariectomy group (OVX) group, and those in the OVX+AG group were treated with AG (100 mg/kg/day) for 8 weeks. Learning and memory were evaluated using the electric Y maze. AGE and Aβ1-40 biochemical assessments were performed using enzyme-linked immunosorbent assay (ELISA) kits. Furthermore, evaluations of brain amyloid precursor protein 695 (APP695) mRNA expression by RT-PCR and AGE expression by immunohistochemistry were carried out. Ovariectomized rats exhibited memory impairment and Aβ production disorder with upregulated APP695 mRNA and AGE expression levels. AG pretreatment relieved the ovariectomy-induced learning and memory disorder and significantly ameliorated the Aβ production disturbance and AGE generation. Additionally, pathological changes in morphology were also significantly recovered. Our data reveal that AG plays a potentially neuroprotective role against ovariectomy-induced learning and cognitive impairment and Aβ production disorder. Advanced glycation end products (AGEs) have been reported to cause neurodegeneration, senile plaque formation and spatial learning and memory deficits. There is much evidence describing the beneficial effects of aminoguanidine (AG) on the central nervous system; AG is able to inhibit the receptor for AGEs and beta-amyloid (Aβ) deposition in the brain, thus preventing cognitive decline and neurodegeneration. In this study, we investigated whether AG protects against ovariectomy-induced neuronal deficits and Aβ deposition in rats. Animals in the ovariectomy group (OVX) group, and those in the OVX+AG group were treated with AG (100 mg/kg/day) for 8 weeks. Learning and memory were evaluated using the electric Y maze. AGE and Aβ1-40 biochemical assessments were performed using enzyme-linked immunosorbent assay (ELISA) kits. Furthermore, evaluations of brain amyloid precursor protein 695 (APP695) mRNA expression by RT-PCR and AGE expression by immunohistochemistry were carried out. Ovariectomized rats exhibited memory impairment and Aβ production disorder with upregulated APP695 mRNA and AGE expression levels. AG pretreatment relieved the ovariectomy-induced learning and memory disorder and significantly ameliorated the Aβ production disturbance and AGE generation. Additionally, pathological changes in morphology were also significantly recovered. Our data reveal that AG plays a potentially neuroprotective role against ovariectomy-induced learning and cognitive impairment and Aβ production disorder.
Collapse
Affiliation(s)
- Dan Di Zhang
- Department of Geriatric Medicine , Beijing Luhe Hospital Affiliated to Capital Medical University , Beijing , China
| | - Yan Gang Wang
- Department of Gastroenterology , Hebei Provincial Hospital of Traditional Chinese Medicine , Hebei , China
| | - Chun Yan Liu
- Department of Rheumatology , The Third Hospital of Hebei Medical University , Hebei , China
| | - Ze Hou Wang
- Department of Chinese Medicine , Beijing University of Chinese Medicine , Beijing , China
| | - Yue Fen Wang
- Department of Nephropathy , Beijing Hospital of Traditional Chinese Medicine , Capital Medical University , Beijing , China
| |
Collapse
|
17
|
Taxier LR, Gross KS, Frick KM. Oestradiol as a neuromodulator of learning and memory. Nat Rev Neurosci 2020; 21:535-550. [PMID: 32879508 PMCID: PMC8302223 DOI: 10.1038/s41583-020-0362-7] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2020] [Indexed: 12/24/2022]
Abstract
Although hormones such as glucocorticoids have been broadly accepted in recent decades as general neuromodulators of memory processes, sex steroid hormones such as the potent oestrogen 17β-oestradiol have been less well recognized by the scientific community in this capacity. The predominance of females in studies of oestradiol and memory and the general (but erroneous) perception that oestrogens are 'female' hormones have probably prevented oestradiol from being more widely considered as a key memory modulator in both sexes. Indeed, although considerable evidence supports a crucial role for oestradiol in regulating learning and memory in females, a growing body of literature indicates a similar role in males. This Review discusses the mechanisms of oestradiol signalling and provides an overview of the effects of oestradiol on spatial, object recognition, social and fear memories. Although the primary focus is on data collected in females, effects of oestradiol on memory in males will be discussed, as will sex differences in the molecular mechanisms that regulate oestrogenic modulation of memory, which may have important implications for the development of future cognitive therapeutics.
Collapse
Affiliation(s)
- Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| |
Collapse
|
18
|
Schwabe MR, Taxier LR, Frick KM. It takes a neural village: Circuit-based approaches for estrogenic regulation of episodic memory. Front Neuroendocrinol 2020; 59:100860. [PMID: 32781195 PMCID: PMC7669700 DOI: 10.1016/j.yfrne.2020.100860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
Cognitive behaviors, such as episodic memory formation, are complex processes involving coordinated activity in multiple brain regions. However, much of the research on hormonal regulation of cognition focuses on manipulation of one region at a time or provides a single snapshot of how a systemic treatment affects multiple brain regions without investigating how these regions might interact to mediate hormone effects. Here, we use estrogenic regulation of episodic memory as an example of how circuit-based approaches may be incorporated into future studies of hormones and cognition. We first review basic episodic memory circuitry, rapid mechanisms by which 17β-estradiol can alter circuit activity, and current knowledge about 17β-estradiol's effects on episodic memory. Next, we outline approaches that researchers can employ to consider circuit effects in their estrogen research and provide examples of how these methods have been used to examine hormonal regulation of memory and other behaviors.
Collapse
Affiliation(s)
- Miranda R Schwabe
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Lisa R Taxier
- 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
|
19
|
Creighton SD, Stefanelli G, Reda A, Zovkic IB. Epigenetic Mechanisms of Learning and Memory: Implications for Aging. Int J Mol Sci 2020; 21:E6918. [PMID: 32967185 PMCID: PMC7554829 DOI: 10.3390/ijms21186918] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
The neuronal epigenome is highly sensitive to external events and its function is vital for producing stable behavioral outcomes, such as the formation of long-lasting memories. The importance of epigenetic regulation in memory is now well established and growing evidence points to altered epigenome function in the aging brain as a contributing factor to age-related memory decline. In this review, we first summarize the typical role of epigenetic factors in memory processing in a healthy young brain, then discuss the aspects of this system that are altered with aging. There is general agreement that many epigenetic marks are modified with aging, but there are still substantial inconsistencies in the precise nature of these changes and their link with memory decline. Here, we discuss the potential source of age-related changes in the epigenome and their implications for therapeutic intervention in age-related cognitive decline.
Collapse
Affiliation(s)
- Samantha D. Creighton
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (S.D.C.); (G.S.)
| | - Gilda Stefanelli
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (S.D.C.); (G.S.)
| | - Anas Reda
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S, Canada;
| | - Iva B. Zovkic
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (S.D.C.); (G.S.)
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S, Canada;
| |
Collapse
|
20
|
Ramzan F, Baumbach J, Monks AD, Zovkic IB. Histone H2A.Z is required for androgen receptor-mediated effects on fear memory. Neurobiol Learn Mem 2020; 175:107311. [PMID: 32916283 DOI: 10.1016/j.nlm.2020.107311] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/13/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022]
Abstract
Epigenetic factors translate environmental signals into stable outcomes, but how they are influenced by regulators of plasticity remain unclear. We previously showed that androgen receptor overexpression inhibited fear memory in male mice and increased expression of the histone variant H2A.Z, a novel epigenetic regulator of memory. Here, we used conditional-inducible H2A.Z knockout mice to investigate how H2A.Z deletion influences androgenic regulation of fear memory. We showed that conditional inducible H2A.Z deletion blocked memory-enhancing effects of androgen depletion (induced by gonadectomy), and of pharmacological inhibition of the androgen receptor with flutamide. Similarly, H2A.Z deletion blocked the memory-reducing effects of DHT, and DHT treatment in cultured hippocampal neurons altered H2A.Z binding, suggesting that AR is an H2A.Z regulator in neurons. Overall, these data show that fear memory formation is regulated by interactions between sex hormones and epigenetic factors, which has implications for sex differences in fear-related disorders.
Collapse
Affiliation(s)
- Firyal Ramzan
- University of Toronto Mississauga, Department of Psychology, Mississauga, Ontario L5L 1C6, Canada
| | - Jennet Baumbach
- University of Toronto Mississauga, Department of Psychology, Mississauga, Ontario L5L 1C6, Canada
| | - Ashley D Monks
- University of Toronto Mississauga, Department of Psychology, Mississauga, Ontario L5L 1C6, Canada
| | - Iva B Zovkic
- University of Toronto Mississauga, Department of Psychology, Mississauga, Ontario L5L 1C6, Canada.
| |
Collapse
|
21
|
Kovács T, Szabó-Meleg E, Ábrahám IM. Estradiol-Induced Epigenetically Mediated Mechanisms and Regulation of Gene Expression. Int J Mol Sci 2020; 21:ijms21093177. [PMID: 32365920 PMCID: PMC7246826 DOI: 10.3390/ijms21093177] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 12/20/2022] Open
Abstract
Gonadal hormone 17β-estradiol (E2) and its receptors are key regulators of gene transcription by binding to estrogen responsive elements in the genome. Besides the classical genomic action, E2 regulates gene transcription via the modification of epigenetic marks on DNA and histone proteins. Depending on the reaction partner, liganded estrogen receptor (ER) promotes DNA methylation at the promoter or enhancer regions. In addition, ERs are important regulators of passive and active DNA demethylation. Furthermore, ERs cooperating with different histone modifying enzymes and chromatin remodeling complexes alter gene transcription. In this review, we survey the basic mechanisms and interactions between estrogen receptors and DNA methylation, demethylation and histone modification processes as well as chromatin remodeling complexes. The particular relevance of these mechanisms to physiological processes in memory formation, embryonic development, spermatogenesis and aging as well as in pathophysiological changes in carcinogenesis is also discussed.
Collapse
Affiliation(s)
- Tamás Kovács
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pécs, Hungary;
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary;
| | - István M. Ábrahám
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pécs, Hungary;
- Correspondence:
| |
Collapse
|
22
|
The medial prefrontal cortex - hippocampus circuit that integrates information of object, place and time to construct episodic memory in rodents: Behavioral, anatomical and neurochemical properties. Neurosci Biobehav Rev 2020; 113:373-407. [PMID: 32298711 DOI: 10.1016/j.neubiorev.2020.04.007] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/25/2020] [Accepted: 04/06/2020] [Indexed: 12/31/2022]
Abstract
Rats and mice have been demonstrated to show episodic-like memory, a prototype of episodic memory, as defined by an integrated memory of the experience of an object or event, in a particular place and time. Such memory can be assessed via the use of spontaneous object exploration paradigms, variably designed to measure memory for object, place, temporal order and object-location inter-relationships. We review the methodological properties of these tests, the neurobiology about time and discuss the evidence for the involvement of the medial prefrontal cortex (mPFC), entorhinal cortex (EC) and hippocampus, with respect to their anatomy, neurotransmitter systems and functional circuits. The systematic analysis suggests that a specific circuit between the mPFC, lateral EC and hippocampus encodes the information for event, place and time of occurrence into the complex episodic-like memory, as a top-down regulation from the mPFC onto the hippocampus. This circuit can be distinguished from the neuronal component memory systems for processing the individual information of object, time and place.
Collapse
|
23
|
Louwies T, Greenwood-Van Meerveld B. Sex differences in the epigenetic regulation of chronic visceral pain following unpredictable early life stress. Neurogastroenterol Motil 2020; 32:e13751. [PMID: 31667916 PMCID: PMC8628638 DOI: 10.1111/nmo.13751] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/11/2019] [Accepted: 09/28/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND We previously reported that early life stress (ELS) dysregulated glucocorticoid receptor (GR) and corticotrophin-releasing hormone (CRH) expression in the central nucleus of the amygdala (CeA). Epigenetic modifications serve as memories of adverse events that occurred during early life. Therefore, we hypothesized that epigenetic mechanisms alter GR and CRH expression in the CeA and underlie chronic visceral pain after ELS. METHODS Neonatal rats were exposed to unpredictable, predictable ELS, or odor only (no stress control) from postnatal days 8 to 12. In adulthood, visceral sensitivity was assessed or the CeA was isolated for Western blot or ChiP-qPCR to study histone modifications at the GR and CRH promoters. Female adult rats underwent stereotaxic implantation of indwelling cannulas for microinjections of garcinol (HAT inhibitor) into the CeA. After 7 days of microinjections, visceral sensitivity was assessed or the CeA was isolated for ChIP-qPCR assays. RESULTS Unpredictable ELS increased visceral sensitivity in adult female rats, but not in male counterparts. ELS increased histone 3 lysine 9 (H3K9) acetylation in the CeA and H3K9 acetylation levels at the GR promoter in the CeA of adult female rats. After unpredictable ELS, H3K9 acetylation was increased and GR binding was decreased at the CRH promoter. Administration of garcinol in the CeA of adult females, that underwent unpredictable ELS, normalized H3K9 acetylation and restored GR binding at the CRH promoter. CONCLUSION Dysregulated histone acetylation and GR binding at the CRH promoter in the CeA are an important mechanism for "memorizing" ELS events mediating visceral pain in adulthood.
Collapse
Affiliation(s)
- Tijs Louwies
- Oklahoma Center for Neuroscience, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Beverley Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Science Center, Oklahoma City, OK, USA,Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA,VA Medical Center, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| |
Collapse
|
24
|
Baumbach JL, Zovkic IB. Hormone-epigenome interactions in behavioural regulation. Horm Behav 2020; 118:104680. [PMID: 31927018 DOI: 10.1016/j.yhbeh.2020.104680] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 02/06/2023]
Abstract
Interactions between hormones and epigenetic factors are key regulators of behaviour, but the mechanisms that underlie their effects are complex. Epigenetic factors can modify sensitivity to hormones by altering hormone receptor expression, and hormones can regulate epigenetic factors by recruiting epigenetic regulators to DNA. The bidirectional nature of this relationship is becoming increasingly evident and suggests that the ability of hormones to regulate certain forms of behaviour may depend on their ability to induce changes in the epigenome. Moreover, sex differences have been reported for several epigenetic modifications, and epigenetic factors are thought to regulate sexual differentiation of behaviour, although specific mechanisms remain to be understood. Indeed, hormone-epigenome interactions are highly complex and involve both canonical and non-canonical regulatory pathways that may permit for highly specific gene regulation to promote variable forms of behavioural adaptation.
Collapse
Affiliation(s)
- Jennet L Baumbach
- Department of Psychology, University of Toronto Mississauga, Mississauga, Canada
| | - Iva B Zovkic
- Department of Psychology, University of Toronto Mississauga, Mississauga, Canada.
| |
Collapse
|
25
|
Finney CA, Shvetcov A, Westbrook RF, Jones NM, Morris MJ. The role of hippocampal estradiol in synaptic plasticity and memory: A systematic review. Front Neuroendocrinol 2020; 56:100818. [PMID: 31843506 DOI: 10.1016/j.yfrne.2019.100818] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/29/2019] [Accepted: 12/11/2019] [Indexed: 12/31/2022]
Abstract
The consolidation of long-term memory is influenced by various neuromodulators. One of these is estradiol, a steroid hormone that is synthesized both in peripheral endocrine tissue and in the brain, including the hippocampus. Here, we examine the evidence regarding the role of estradiol in the hippocampus, specifically, in memory formation and its effects on the molecular mechanisms underlying synaptic plasticity. We conclude that estradiol improves memory consolidation and, thereby, long-term memory. Previous studies have shown that it does this in three, interconnected ways: (1) via functional changes in excitatory activity, (2) signaling changes in calcium dynamics, protein phosphorylation and protein expression, and (3) structural changes to synaptic morphology. Through a functional network analysis of proteins affected by estradiol, we identify potential protein-protein interactions that further support a role for estradiol in modulating synaptic plasticity as well as highlight signaling pathways that may be involved in these changes within the hippocampus.
Collapse
Affiliation(s)
- C A Finney
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - A Shvetcov
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - R F Westbrook
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - N M Jones
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - M J Morris
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
26
|
Kim EJ, Kim JJ. Amygdala, Medial Prefrontal Cortex and Glucocorticoid Interactions Produce Stress-Like Effects on Memory. Front Behav Neurosci 2019; 13:210. [PMID: 31619974 PMCID: PMC6759673 DOI: 10.3389/fnbeh.2019.00210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/29/2019] [Indexed: 11/17/2022] Open
Abstract
Adverse stress effects on the hippocampal memory system are generally thought to be due to the high level of circulating glucocorticoids directly modifying the properties of hippocampal neurons and, accordingly, the results should be reproducible with exogenous administration of cortisol in humans and corticosterone in rodents. However, glucocorticoid levels increased to other events, such as exercise and environment enrichment, do not impair but instead enhance hippocampal memory, indicating that cortisol/corticosterone are not invariant causal factors of stress. To better model the complex psychophysiological attributes of stress (i.e., aversiveness, lack of controllability, and glucose metabolism), we examined the functions of the amygdala, medial prefrontal cortex (mPFC), and corticosterone on a hippocampal-based one-trial novel object recognition (OR) memory task in rats. Specifically, animals were subjected to amygdala stimulation, mPFC inactivation, and corticosterone treatments separately or in combination during behavioral testing. Collective amygdala, mPFC, and corticosterone manipulations significantly impaired OR memory comparable to behavioral stress. By contrast, single and dual treatments failed to reliably decrease memory functioning. These results suggest that negative mnemonic impacts of uncontrollable stress involve the amalgamation of heightened amygdala and diminished mPFC activities, and elevated circulating corticosterone level.
Collapse
Affiliation(s)
- Eun Joo Kim
- Department of Psychology, University of Washington, Seattle, WA, United States
| | - Jeansok J Kim
- Department of Psychology, University of Washington, Seattle, WA, United States.,Program in Neuroscience, University of Washington, Seattle, WA, United States
| |
Collapse
|
27
|
S-nitrosoglutathione prevents cognitive impairment through epigenetic reprogramming in ovariectomised mice. Biochem Pharmacol 2019; 168:352-365. [DOI: 10.1016/j.bcp.2019.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/23/2019] [Indexed: 12/22/2022]
|
28
|
Keiser AA, Wood MA. Examining the contribution of histone modification to sex differences in learning and memory. Learn Mem 2019; 26:318-331. [PMID: 31416905 PMCID: PMC6699407 DOI: 10.1101/lm.048850.118] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/08/2019] [Indexed: 01/04/2023]
Abstract
The epigenome serves as a signal integration platform that encodes information from experience and environment that adds tremendous complexity to the regulation of transcription required for memory, beyond the directions encoded in the genome. To date, our understanding of how epigenetic mechanisms integrate information to regulate gene expression required for memory is primarily obtained from male derived data despite sex-specific life experiences and sex differences in consolidation and retrieval of memory, and in the molecular mechanisms that mediate these processes. In this review, we examine the contribution of chromatin modification to learning and memory in both sexes. We provide examples of how exposure to a number of internal and external factors influence the epigenome in sex-similar and sex-specific ways that may ultimately impact transcription required for memory processes. We also pose a number of key open questions and identify areas requiring further investigation as we seek to understand how histone modifying mechanisms shape memory in females.
Collapse
Affiliation(s)
- Ashley A Keiser
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, USA
| |
Collapse
|
29
|
Taxier LR, Philippi SM, Fortress AM, Frick KM. Dickkopf-1 blocks 17β-estradiol-enhanced object memory consolidation in ovariectomized female mice. Horm Behav 2019; 114:104545. [PMID: 31228421 PMCID: PMC6732224 DOI: 10.1016/j.yhbeh.2019.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/15/2019] [Accepted: 06/16/2019] [Indexed: 01/05/2023]
Abstract
The memory-enhancing effects of 17β-estradiol (E2) depend upon rapid activation of several cell-signaling cascades within the dorsal hippocampus (DH). Among the many cell-signaling pathways that mediate memory processes, Wnt/β-catenin signaling has emerged as a potential key player because of its importance to hippocampal development and synaptic plasticity. However, whether E2 interacts with Wnt/β-catenin signaling to promote memory consolidation is unknown. Therefore, the present study examined whether Wnt/β-catenin signaling within the DH is necessary for E2-induced memory consolidation in ovariectomized mice tested in the object recognition and object placement tasks. Ovariectomized C57BL/6 mice received immediate post-training infusions of E2 or vehicle into the dorsal third ventricle plus the endogenous Wnt/β-catenin antagonist Dickkopf-1 (Dkk-1) or vehicle into the DH to assess whether the memory-enhancing effects of E2 depend on activation of Wnt/β-catenin signaling. Our results suggest that Dkk-1 blocks E2-induced memory enhancement as hypothesized, but may do so by only moderately blunting Wnt/β-catenin signaling while concurrently activating Wnt/JNK signaling. The current study provides novel insights into the mechanisms through which E2 enhances memory consolidation in the DH, as well as critical information about the mechanistic actions of Dkk-1.
Collapse
Affiliation(s)
- Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| | - Sarah M Philippi
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| | - Ashley M Fortress
- V.A. Pittsburgh Healthcare System, Pittsburgh, PA 15216, United States.
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
30
|
Koss WA, Frick KM. Activation of androgen receptors protects intact male mice from memory impairments caused by aromatase inhibition. Horm Behav 2019; 111:96-104. [PMID: 30653980 PMCID: PMC6527464 DOI: 10.1016/j.yhbeh.2019.01.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/21/2018] [Accepted: 01/10/2019] [Indexed: 12/17/2022]
Abstract
Although 17β-estradiol (E2) is known to regulate hippocampal function, the specific contributions of hippocampally-synthesized E2 remain unclear. Infusion of the aromatase inhibitor letrozole into the dorsal hippocampus (DH) of ovariectomized mice disrupts object recognition and object placement memory consolidation, suggesting that DH-synthesized E2 is essential for memory. However, the role of DH-synthesized E2 in memory among male rodents is unknown. Here, we examined effects of aromatase inhibition on memory consolidation in male mice. Intact and gonadectomized mice were infused with vehicle or letrozole into the DH immediately post-training in object placement and object recognition tasks. Letrozole blocked memory in both tasks among gonadectomized males only, suggesting that circulating androgens, or a rise in hippocampal androgens due to aromatase inhibition, may support memory consolidation in intact males. To test this hypothesis, intact males were infused with the androgen receptor antagonist flutamide into the DH after object training. A dose-dependent impairment was observed in both tasks, indicating that blocking androgen signaling can impair memory consolidation. To test if hippocampal androgen receptor activation protected intact males from the impairing effects of letrozole, a non-impairing dose of flutamide was co-infused with letrozole. Co-administration of both drugs blocked object placement and object recognition memory consolidation, demonstrating that letrozole impairs memory in intact males only if androgen receptors are blocked. Together, these data suggest that DH-synthesized E2 and androgen receptor activation may work in concert to mediate memory consolidation in intact males, such that androgen receptor activation protects against memory impairments caused by aromatase inhibition.
Collapse
Affiliation(s)
- Wendy A Koss
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States of America.
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States of America.
| |
Collapse
|
31
|
Deb S, Phukan BC, Mazumder MK, Dutta A, Paul R, Bhattacharya P, Sandhir R, Borah A. Garcinol, a multifaceted sword for the treatment of Parkinson's disease. Neurochem Int 2019; 128:50-57. [PMID: 30986504 DOI: 10.1016/j.neuint.2019.04.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/23/2022]
Abstract
Garcinol, the principal phytoconstituent of plants belonging to the genus Garcinia, is known for its anti-oxidant as well as anti-inflammatory properties, which can be extended to its possible neuroprotective role. Recent reports disseminate the capacity of garcinol to influence neuronal growth and survival, alter the neurochemical status in brain, as well as regulate memory and cognition. The concomitant neuro-rescue property of garcinol may render it as an effective compound in Parkinson's disease (PD) therapeutics since it is capable of ameliorating the related pathophysiological changes. Emerging pieces of evidence linking histone acetylation defects to the progression of neurodegenerative diseases provide an effective basis for targeting PD. Hyperacetylation of histones has been reported in Parkinsonian brain, which demands the use of pharmacological inhibitors of histone acetyltransferases (HAT). Garcinol serves as a potent natural HAT inhibitor and has unveiled promising results in molecular interaction studies against Monoamine oxidase B (MAO-B) and Catechol-O-Methyltransferase (COMT), as well as in L-DOPA induced dyskinesia. This review highlights the prospective implications of garcinol as a novel anti-Parkinsonian agent, and establishes a bridge between histone acetylation defects and the pathological aspects of PD.
Collapse
Affiliation(s)
- Satarupa Deb
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India
| | - Banashree Chetia Phukan
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India
| | - Muhammed Khairujjaman Mazumder
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India
| | - Ankumoni Dutta
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India
| | - Rajib Paul
- Department of Zoology, Pandit Deendayal Upadhyaya Adarsha Mahavidyalaya (PDUAM), Eraligool, 788723, Karimganj, Assam, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, 382355, Gandhinagar, Gujarat, India
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, 788011, Assam, India.
| |
Collapse
|
32
|
Suberoylanilide hydroxamic acid reversed cognitive and synaptic plasticity impairments induced by sevoflurane exposure in adult mice. Neuroreport 2019; 30:274-279. [DOI: 10.1097/wnr.0000000000001196] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
33
|
Sex differences and the neurobiology of affective disorders. Neuropsychopharmacology 2019; 44:111-128. [PMID: 30061743 PMCID: PMC6235863 DOI: 10.1038/s41386-018-0148-z] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/14/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
Abstract
Observations of the disproportionate incidence of depression in women compared with men have long preceded the recent explosion of interest in sex differences. Nonetheless, the source and implications of this epidemiologic sex difference remain unclear, as does the practical significance of the multitude of sex differences that have been reported in brain structure and function. In this article, we attempt to provide a framework for thinking about how sex and reproductive hormones (particularly estradiol as an example) might contribute to affective illness. After briefly reviewing some observed sex differences in depression, we discuss how sex might alter brain function through hormonal effects (both organizational (programmed) and activational (acute)), sex chromosome effects, and the interaction of sex with the environment. We next review sex differences in the brain at the structural, cellular, and network levels. We then focus on how sex and reproductive hormones regulate systems implicated in the pathophysiology of depression, including neuroplasticity, genetic and neural networks, the stress axis, and immune function. Finally, we suggest several models that might explain a sex-dependent differential regulation of affect and susceptibility to affective illness. As a disclaimer, the studies cited in this review are not intended to be comprehensive but rather serve as examples of the multitude of levels at which sex and reproductive hormones regulate brain structure and function. As such and despite our current ignorance regarding both the ontogeny of affective illness and the impact of sex on that ontogeny, sex differences may provide a lens through which we may better view the mechanisms underlying affective regulation and dysfunction.
Collapse
|
34
|
Zhang Y, Dong HT, Duan L, Niu L, Yuan GQ, Dai JQ, Hou BR, Pan YW. HDAC4 gene silencing alleviates epilepsy by inhibition of GABA in a rat model. Neuropsychiatr Dis Treat 2019; 15:405-416. [PMID: 30787615 PMCID: PMC6366349 DOI: 10.2147/ndt.s181669] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES Despite the availability of effective antiepileptic drugs, epileptic patients still suffer from intractable seizures and adverse events. Better control of both seizures and fewer side effects is needed in order to enhance the patient's quality of life. We performed the present study with an attempt to explore the effect that HDAC4 gene silencing would have on epilepsy simulated by model rats. Furthermore, the study made additional analysis on the relativity of the HDAC4 gene in regard to its relationship with the gamma-aminobutyric acid (GABA) signaling pathway. MATERIALS AND METHODS Tremor rats were prepared in order to establish the epilepsy model. The rats would go on to be treated with si-HDAC4 in order to identify roles of the HDAC4 in levels of GABAARα1, GABAARα4, GAD65, GAT-1, and GAT-3. Finally, both electroencephalogram behavior and cognitive function of the rats following the treatment of si-HDAC4 were observed. RESULTS Levels of the GABAARα1 and GABAARα4 showed an evident increase, while GAD65, GAT-1, and GAT-3 displayed a decline in the epilepsy rats treated with the aforementioned si-HDAC4 when compared with the epilepsy rats. After injection of si-HDAC4, the epilepsy rats presented with a reduction in seizure degree, latency and duration of seizure, amount of scattered epileptic waves, and occurrence of epilepsy, with an improvement in their cognitive function. CONCLUSION The study highlighted the role that HDAC4 gene silencing played in easing the cases of epilepsy found in the model rats. This was shown to have occurred through the upregulation of both GABAARα1 and GABAARα4 levels, as well as in the downregulation of GAD65, GAT-1, and GAT-3 levels. The evidence provided shows that the HDAC4 gene is likely to present as a new objective in further experimentation in the treatment of epilepsy.
Collapse
Affiliation(s)
- Yinian Zhang
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, People's Republic of China, .,Institute of Neurology, Lanzhou University, Lanzhou 730030, People's Republic of China,
| | - Hua-Teng Dong
- Department of Pediatric Neurology, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou 730050, People's Republic of China
| | - Lei Duan
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, People's Republic of China, .,Institute of Neurology, Lanzhou University, Lanzhou 730030, People's Republic of China,
| | - Liang Niu
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, People's Republic of China, .,Institute of Neurology, Lanzhou University, Lanzhou 730030, People's Republic of China,
| | - Guo-Qiang Yuan
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, People's Republic of China, .,Institute of Neurology, Lanzhou University, Lanzhou 730030, People's Republic of China,
| | - Jun-Qiang Dai
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, People's Republic of China, .,Institute of Neurology, Lanzhou University, Lanzhou 730030, People's Republic of China,
| | - Bo-Ru Hou
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, People's Republic of China, .,Institute of Neurology, Lanzhou University, Lanzhou 730030, People's Republic of China,
| | - Ya-Wen Pan
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, People's Republic of China, .,Institute of Neurology, Lanzhou University, Lanzhou 730030, People's Republic of China,
| |
Collapse
|
35
|
Chemogenetic inactivation of the dorsal hippocampus and medial prefrontal cortex, individually and concurrently, impairs object recognition and spatial memory consolidation in female mice. Neurobiol Learn Mem 2018; 156:103-116. [PMID: 30408525 PMCID: PMC7310386 DOI: 10.1016/j.nlm.2018.11.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/25/2018] [Accepted: 11/03/2018] [Indexed: 01/23/2023]
Abstract
The dorsal hippocampus (DH) and medial prefrontal cortex (mPFC) are brain regions essential for processing and storing episodic memory. In rodents, the DH has a well-established role in supporting the consolidation of episodic-like memory in tasks such as object recognition and object placement. However, the role of the mPFC in the consolidation of episodic-like memory tasks remains controversial. Therefore, the present study examined involvement of the DH and mPFC, alone and in combination, in object and spatial recognition memory consolidation in ovariectomized female mice. To this end, we utilized two types of inhibitory Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) to inactivate the DH alone, the mPFC alone, or both brain regions concurrently immediately after object training to assess the role of each region in the consolidation of object recognition and spatial memories. Our results using single and multiplexed DREADDS suggest that excitatory activity in the DH and mPFC, alone or in combination, is required for the successful consolidation of object recognition and spatial memories. Together, these studies provide critical insight into how the DH and mPFC work in concert to facilitate memory consolidation in female mice.
Collapse
|
36
|
Sex Differences in the Rapid Cell Signaling Mechanisms Underlying the Memory-Enhancing Effects of 17β-Estradiol. eNeuro 2018; 5:eN-NWR-0267-18. [PMID: 30406188 PMCID: PMC6220582 DOI: 10.1523/eneuro.0267-18.2018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/07/2018] [Accepted: 09/30/2018] [Indexed: 12/20/2022] Open
Abstract
Little is known about how 17β-estradiol (E2) mediates memory formation in males. In ovariectomized (OVX) mice, bilateral dorsal hippocampal (DH) infusion of E2 enhances memory consolidation in object recognition (OR) and object placement (OP) tasks in a manner dependent on activation of extracellular signal-regulated kinase (ERK) and Akt signaling. Here, bilateral DH E2 infusion enhanced memory consolidation in both tasks among OVX female, gonadally-intact male, and castrated male mice, suggesting comparable facilitation of memory consolidation in both sexes, independent of testicular hormones in males. Contrary to previous reports in OVX mice, E2 did not increase DH ERK or Akt phosphorylation in males, nor did the ERK inhibitor U0126 [1,4-diamino-2,3-dicyano-1,4-bis (o-aminophenylmercapto) butadiene] prevent E2 from enhancing memory consolidation among intact and castrated males. These data suggest that ERK activation is not necessary for E2 to enhance memory consolidation in males, and compared with previous reports in females, reveal novel sex differences in the cell-signaling pathways through which E2 facilitates memory consolidation. To explore the mechanisms underlying E2-induced memory enhancements in males, phosphorylation of the transcription factor cAMP response element binding protein (CREB) in the DH was assessed. E2 increased phospho-CREB levels in both sexes, yet U0126 did not block these increases in castrated or intact males, indicating that E2 regulates CREB phosphorylation in males via an ERK-independent mechanism. Collectively, these findings suggest that the beneficial effects of hippocampal E2 on memory consolidation in males and females are mediated by different molecular mechanisms, which has important implications for the development of treatments to reduce memory dysfunction in men and women.
Collapse
|
37
|
Sellers KJ, Watson IA, Gresz RE, Raval P, Srivastava DP. Cyto-nuclear shuttling of afadin is required for rapid estradiol-mediated modifications of histone H3. Neuropharmacology 2018; 143:153-162. [PMID: 30268521 PMCID: PMC6277849 DOI: 10.1016/j.neuropharm.2018.09.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/18/2018] [Accepted: 09/25/2018] [Indexed: 12/19/2022]
Abstract
Estrogens have been shown to rapidly regulate local signalling at synapses and within the nucleus. The result of these signalling events is to rapidly modulate synapse structure and function, as well as epigenetic mechanisms including histone modifications. Ultimately these mechanisms are thought to contribute to long-lasting changes in neural circuitry, and thus influence cognitive functions such as learning and memory. However, the mechanisms by which estrogen-mediated local synaptic and nuclear signalling events are coordinated are not well understood. In this study we have found that the scaffold protein afadin, (also known as AF-6), undergoes a bi-directional trafficking to both synaptic and nuclear compartment in response to acute 17β-estradiol (estradiol) treatment, in mixed sex neuronal cultures derived from fetal cortex. Interestingly, nuclear accumulation of afadin was coincidental with an increase in the phosphorylation of histone H3 at serine 10 (H3S10p). This epigenetic modification is associated with the remodeling of chromatin into an open euchromatin state, allowing for transcriptional activation and related learning and memory processes. Critically, the cyto-nuclear trafficking of afadin was required for estradiol-dependent H3S10p. We further determined that nuclear accumulation of afadin is sufficient to induce phosphorylation of the mitogentic kinases ERK1/2 (pERK1/2) within the nucleus. Moreover, nuclear pERK1/2 was required for estradiol-dependent H3S10p. Taken together, we propose a model whereby estradiol induces the bi-directional trafficking of afadin to synaptic and nuclear sub-compartments. Within the nucleus, afadin is required for increased pERK1/2 which in turn is required for H3S10p. Therefore this represents a mechanism through which estrogens may be able to coordinate both synaptic and nucleosomal events within the same neuronal population. 17β-estradiol targets afadin to membrane and nuclear subcompartments. Histone H3 is rapidly phosphorylated by 17β-estradiol. Histone H3 phosphorylation by 17β-estradiol requires afadin nuclear accumulation. 17β-estradiol-mediated ERK1/2 activation is required for histone H3 phosphorylation.
Collapse
Affiliation(s)
- Katherine J Sellers
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK
| | - Iain A Watson
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK
| | - Rahel E Gresz
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK
| | - Pooja Raval
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE5 9RT, UK.
| |
Collapse
|
38
|
Frick KM, Kim J. Mechanisms underlying the rapid effects of estradiol and progesterone on hippocampal memory consolidation in female rodents. Horm Behav 2018; 104:100-110. [PMID: 29727606 PMCID: PMC6226372 DOI: 10.1016/j.yhbeh.2018.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 12/21/2022]
Abstract
Contribution to Special Issue on Fast effects of steroids. Although rapid effects of 17β‑estradiol (E2) and progesterone on cellular functions have been observed for several decades, a proliferation of data in recent years has demonstrated the importance of these actions to cognition. In particular, an emerging literature has demonstrated that these hormones promote the consolidation of spatial and object recognition memories in rodents via rapid activation of numerous cellular events including cell signaling, histone modifications, and local protein translation in the hippocampus. This article provides an overview of the evidence demonstrating that E2 and progesterone enhance hippocampal memory consolidation in female rodents, and then discusses numerous molecular mechanisms thus far shown to mediate the beneficial effects of these hormones on memory formation.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| | - Jaekyoon Kim
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| |
Collapse
|
39
|
Djiogue S, Djiyou Djeuda AB, Seke Etet PF, Ketcha Wanda GJM, Djikem Tadah RN, Njamen D. Memory and exploratory behavior impairment in ovariectomized Wistar rats. Behav Brain Funct 2018; 14:14. [PMID: 30012162 PMCID: PMC6047120 DOI: 10.1186/s12993-018-0146-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/10/2018] [Indexed: 01/01/2023] Open
Abstract
Background Estrogen deficiency is linked to changes in several physiological processes, but the extent to which it associates with cognitive changes in menopause context is controversial. Rationale We evaluated the impact of ovariectomy on memory processes and normal exploratory behavior in Wistar rats. Methods Young adult rats (4–6 months) were either ovariectomized (OVX group) (N = 10), sham operated (N = 10), or untouched (naïve controls) (N = 8). Afterwards, they were monitored for 12 weeks during which their cognitive functions were evaluated at first week (S1), second (S2), every 3 weeks (S5, S8) and then at week 12 (S12) using: (i) object recognition test to evaluate the short-term and long-term non-spatial memory; (ii) the object placement test to assess the spatial memory; and (iii) normal exploratory behavior components like locomotor and vertical activities in an open field arena. Results Marked changes in ovariectomized rats were observed in long-term non-spatial memory (~ 40% change vs. naïve and sham, P < 0.001) and spatial memory (~ 30% change, P < 0.05) from S2. Instead, from S5 the exploratory behavior was affected, with decreases in line crossing and rearing episode numbers (~ 40% change, P < 0.01), and in the time spent in the center of open field arena (~ 60% change, P < 0.01). Conclusions Our findings support the involvement of sex hormones in cognitive functions in female rats and suggest that controversy on the importance of cognitive affections in menopause context may emerge from differences between short-term and long-term memory processes.
Collapse
Affiliation(s)
- Sefirin Djiogue
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon.
| | - Armando Blondel Djiyou Djeuda
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon
| | - Paul Faustin Seke Etet
- Center for Sustainable Health and Development, University of Ngaoundere, P.O. Box 454, Ngaoundere, Cameroon
| | | | - Rudig Nikanor Djikem Tadah
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon
| | - Dieudonne Njamen
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon
| |
Collapse
|
40
|
Giatti S, Garcia-Segura LM, Barreto GE, Melcangi RC. Neuroactive steroids, neurosteroidogenesis and sex. Prog Neurobiol 2018; 176:1-17. [PMID: 29981391 DOI: 10.1016/j.pneurobio.2018.06.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/25/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022]
Abstract
The nervous system is a target and a source of steroids. Neuroactive steroids are steroids that target neurons and glial cells. They include hormonal steroids originated in the peripheral glands, steroids locally synthesized by the neurons and glial cells (neurosteroids) and synthetic steroids, some of them used in clinical practice. Here we review the mechanisms of synthesis, metabolism and action of neuroactive steroids, including the role of epigenetic modifications and the mitochondria in their sex specific actions. We examine sex differences in neuroactive steroid levels under physiological conditions and their role in the establishment of sex dimorphic structures in the nervous system and sex differences in its function. In addition, particular attention is paid to neuroactive steroids under pathological conditions, analyzing how pathology alters their levels and their role as neuroprotective factors, considering the influence of sex in both cases.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
41
|
Abstract
Estrogens influence nearly every aspect of hippocampal function, including memory formation. Although this research has traditionally focused on ovariectomized females, more recent work is providing insights into the ways in which estrogens regulate hippocampal function in both sexes. This review provides an overview of estrogenic regulation of hippocampal function in female and male rodents, with a particular emphasis on memory formation. Where applicable, we discuss the involvement of specific estrogen receptors and molecular mechanisms that mediate these effects. The review concludes by suggesting gaps in the literature that need to be filled to provide greater insights into potential sex differences in the effects of estrogens on hippocampal function.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211
| | - Jaekyoon Kim
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211
| | - Wendy A Koss
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211
| |
Collapse
|
42
|
Frick KM, Tuscher JJ, Koss WA, Kim J, Taxier LR. Estrogenic regulation of memory consolidation: A look beyond the hippocampus, ovaries, and females. Physiol Behav 2018; 187:57-66. [PMID: 28755863 PMCID: PMC5787049 DOI: 10.1016/j.physbeh.2017.07.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/14/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022]
Abstract
The potent estrogen 17β-estradiol (E2) has long been known to regulate the hippocampus and hippocampal-dependent memories in females, and research from the past decade has begun to shed light on the molecular mechanisms through which E2 mediates memory formation in females. Although E2 can also regulate hippocampal function in males, relatively little is known about how E2 influences memory formation in males, or whether sex differences in underlying mechanisms exist. This review, based on a talk given in April 2017 at the American University symposium entitled, "Sex Differences: From Neuroscience to the Clinic and Beyond", first provides an overview of the molecular mechanisms in the dorsal hippocampus through which E2 enhances memory consolidation in ovariectomized female mice. Next, newer research is described demonstrating key roles for the prefrontal cortex and de novo hippocampal E2 synthesis to the memory-enhancing effects of E2 in females. The review then discusses the effects of de novo and exogenous E2 on hippocampal memory consolidation in both sexes, and putative sex differences in the underlying molecular mechanisms through which E2 enhances memory formation. The review concludes by discussing the importance and implications of sex differences in the molecular mechanisms underlying E2-induced memory consolidation for human health.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| | - Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Wendy A Koss
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Jaekyoon Kim
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| |
Collapse
|
43
|
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
|
44
|
Sheppard PAS, Koss WA, Frick KM, Choleris E. Rapid actions of oestrogens and their receptors on memory acquisition and consolidation in females. J Neuroendocrinol 2018; 30. [PMID: 28489296 PMCID: PMC6543823 DOI: 10.1111/jne.12485] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 12/20/2022]
Abstract
Increased attention has been paid in recent years to the ways in which oestrogens and oestrogen receptors rapidly affect learning and memory. These rapid effects occur within a timeframe that is too narrow for the classical genomic mode of action of oestrogen, thus suggesting nonclassical effects as underlying mechanisms. The present review examines recent developments in the study of the rapid effects of 17β-oestradiol and oestrogen receptor (ER) agonists on learning and memory tasks in female rodents, including social recognition, object recognition, object placement (spatial memory) and social learning. By comparing studies utilising systemic or intracranial treatments, as well as pre- and post-acquisition administration of oestradiol or ER agonists, the respective contributions of individual ERs within specific brain regions to various forms of learning and memory can be determined. The first part of this review explores the effects of systemic administration of 17β-oestradiol and ER agonists on memory when administered either pre- or post-acquisition. The second part not only focuses on the effects of pre- and post-acquisition infusions of 17β-oestradiol or ER agonists into the dorsal hippocampus on memory, but also discusses the contributions of other brain regions, including the medial amygdala, medial prefrontal cortex and paraventricular nucleus of the hypothalamus. The cellular mechanisms mediating the rapid effects of 17β-oestradiol on memory, including activation of intracellular signalling cascades and epigenetic processes, are discussed. Finally, the review concludes by comparing pre- and post-acquisition findings and effects of 17β-oestradiol and ER agonists in different brain regions.
Collapse
Affiliation(s)
- P A S Sheppard
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - W A Koss
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - K M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - E Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
45
|
Singh P, Srivas S, Thakur MK. Epigenetic Regulation of Memory-Therapeutic Potential for Disorders. Curr Neuropharmacol 2017; 15:1208-1221. [PMID: 28393704 PMCID: PMC5725549 DOI: 10.2174/1570159x15666170404144522] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/03/2017] [Accepted: 03/25/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Memory is a vital function which declines in different physiological and pathological conditions such as aging and neurodegenerative diseases. Research in the past has reported that memory formation and consolidation require the precise expression of synaptic plasticity genes. However, little is known about the regulation of these genes. Epigenetic modification is now a well established mechanism that regulates synaptic plasticity genes and neuronal functions including memory. Therefore, we have reviewed the epigenetic regulation of memory and its therapeutic potential for memory dysfunction during aging and neurological disorders. METHOD Research reports and online contents relevant to epigenetic regulation of memory during physiological and pathological conditions have been compiled and discussed. RESULTS Epigenetic modifications include mainly DNA methylation and hydroxymethylation, histone acetylation and methylation which involve chromatin modifying enzymes. These epigenetic marks change during memory formation and impairment due to dementia, aging and neurodegeneration. As the epigenetic modifications are reversible, they can be modulated by enzyme inhibitors leading to the recovery of memory. CONCLUSION Epigenetic modifications could be exploited as a potential therapeutic target to recover memory disorders during aging and pathological conditions.
Collapse
Affiliation(s)
- Padmanabh Singh
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Sweta Srivas
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - M K Thakur
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| |
Collapse
|
46
|
Ratnu VS, Emami MR, Bredy TW. Genetic and epigenetic factors underlying sex differences in the regulation of gene expression in the brain. J Neurosci Res 2017; 95:301-310. [PMID: 27870402 DOI: 10.1002/jnr.23886] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/13/2016] [Accepted: 07/25/2016] [Indexed: 12/14/2022]
Abstract
There are inherent biological differences between males and females that contribute to sex differences in brain function and to many sex-specific illnesses and disorders. Traditionally, it has been thought that such differences are due largely to hormonal regulation; however, there are also genetic and epigenetic effects caused by the inheritance and unequal dosage of genes located on the X and Y chromosomes. Here we discuss the evidence in favor of a genetic and epigenetic basis for sexually dimorphic behavior, as a consequence of underlying differences in the regulation of genes that drive brain function. A better understanding of sex-specific molecular processes in the brain will provide further insight for the development of novel therapeutic approaches for the treatment of neuropsychiatric disorders characterized by sex differences. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vikram S Ratnu
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Michael R Emami
- Department of Neurobiology and Behavior, University of California, Irvine, California
| | - Timothy W Bredy
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurobiology and Behavior, University of California, Irvine, California
| |
Collapse
|
47
|
Abstract
This article highlights the emerging therapeutic potential of specific epigenetic modulators as promising antiepileptogenic or disease-modifying agents for curing epilepsy. Currently, there is an unmet need for antiepileptogenic agents that truly prevent the development of epilepsy in people at risk. There is strong evidence that epigenetic signaling, which exerts high fidelity regulation of gene expression, plays a crucial role in the pathophysiology of epileptogenesis and chronic epilepsy. These modifications are not hard-wired into the genome and are constantly reprogrammed by environmental influences. The potential epigenetic mechanisms, including histone modifications, DNA methylation, microRNA-based transcriptional control, and bromodomain reading activity, can drastically alter the neuronal gene expression profile by exerting their summative effects in a coordinated fashion. Such an epigenetic intervention appears more rational strategy for preventing epilepsy because it targets the primary pathway that initially triggers the numerous downstream cellular and molecular events mediating epileptogenesis. Among currently approved epigenetic drugs, the majority are anticancer drugs with well-established profiles in clinical trials and practice. Evidence from preclinical studies supports the premise that these drugs may be applied to a wide range of brain disorders. Targeting histone deacetylation by inhibiting histone deacetylase enzymes appears to be one promising epigenetic therapy since certain inhibitors have been shown to prevent epileptogenesis in animal models. However, developing neuronal specific epigenetic modulators requires rational, pathophysiology-based optimization to efficiently intercept the upstream pathways in epileptogenesis. Overall, epigenetic agents have been well positioned as new frontier tools towards the national goal of curing epilepsy.
Collapse
Affiliation(s)
- Iyan Younus
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA.
| |
Collapse
|
48
|
Mitchnick KA, Creighton SD, Cloke JM, Wolter M, Zaika O, Christen B, Van Tiggelen M, Kalisch BE, Winters BD. Dissociable roles for histone acetyltransferases p300 and PCAF in hippocampus and perirhinal cortex-mediated object memory. GENES BRAIN AND BEHAVIOR 2017; 15:542-57. [PMID: 27251651 DOI: 10.1111/gbb.12303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/27/2016] [Accepted: 05/29/2016] [Indexed: 10/21/2022]
Abstract
The importance of histone acetylation for certain types of memory is now well established. However, the specific contributions of the various histone acetyltransferases to distinct memory functions remain to be determined; therefore, we employed selective histone acetyltransferase protein inhibitors and short-interference RNAs to evaluate the roles of CREB-binding protein (CBP), E1A-binding protein (p300) and p300/CBP-associated factor (PCAF) in hippocampus and perirhinal cortex (PRh)-mediated object memory. Rats were tested for short- (STM) and long-term memory (LTM) in the object-in-place task, which relies on the hippocampus and PRh for spatial memory and object identity processing, respectively. Selective inhibition of these histone acetyltransferases by small-interfering RNA and pharmacological inhibitors targeting the HAT domain produced dissociable effects. In the hippocampus, CBP or p300 inhibition impaired long-term but not short-term object memory, while inhibition of PCAF impaired memory at both delays. In PRh, HAT inhibition did not impair STM, and only CBP and PCAF inhibition disrupted LTM; p300 inhibition had no effects. Messenger RNA analyses revealed findings consistent with the pattern of behavioral effects, as all three enzymes were upregulated in the hippocampus (dentate gyrus) following learning, whereas only CBP and PCAF were upregulated in PRh. These results demonstrate, for the first time, the necessity of histone acetyltransferase activity for PRh-mediated object memory and indicate that the specific mnemonic roles of distinctive histone acetyltransferases can be dissociated according to specific brain regions and memory timeframe.
Collapse
Affiliation(s)
- K A Mitchnick
- Department of Psychology, University of Guelph, Guelph, ON, Canada.,Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - S D Creighton
- Department of Psychology, University of Guelph, Guelph, ON, Canada.,Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - J M Cloke
- Department of Psychology, University of Guelph, Guelph, ON, Canada.,Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - M Wolter
- Department of Psychology, University of Guelph, Guelph, ON, Canada
| | - O Zaika
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - B Christen
- Department of Psychology, University of Guelph, Guelph, ON, Canada
| | - M Van Tiggelen
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - B E Kalisch
- Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada.,Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - B D Winters
- Department of Psychology, University of Guelph, Guelph, ON, Canada.,Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
49
|
Koss WA, Frick KM. Sex differences in hippocampal function. J Neurosci Res 2016; 95:539-562. [DOI: 10.1002/jnr.23864] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/28/2016] [Accepted: 07/11/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Wendy A. Koss
- Department of Psychology; University of Wisconsin-Milwaukee; Milwaukee Wisconsin
| | - Karyn M. Frick
- Department of Psychology; University of Wisconsin-Milwaukee; Milwaukee Wisconsin
| |
Collapse
|
50
|
Griñan-Ferré C, Puigoriol-Illamola D, Palomera-Ávalos V, Pérez-Cáceres D, Companys-Alemany J, Camins A, Ortuño-Sahagún D, Rodrigo MT, Pallàs M. Environmental Enrichment Modified Epigenetic Mechanisms in SAMP8 Mouse Hippocampus by Reducing Oxidative Stress and Inflammaging and Achieving Neuroprotection. Front Aging Neurosci 2016; 8:241. [PMID: 27803663 PMCID: PMC5067530 DOI: 10.3389/fnagi.2016.00241] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/29/2016] [Indexed: 12/21/2022] Open
Abstract
With the increase in life expectancy, aging and age-related cognitive impairments are becoming one of the most important issues for human health. At the same time, it has been shown that epigenetic mechanisms are emerging as universally important factors in life expectancy. The Senescence Accelerated Mouse P8 (SAMP8) strain exhibits age-related deterioration evidenced in learning and memory abilities and is a useful model of neurodegenerative disease. In SAMP8, Environmental Enrichment (EE) increased DNA-methylation levels (5-mC) and reduced hydroxymethylation levels (5-hmC), as well as increased histone H3 and H4 acetylation levels. Likewise, we found changes in the hippocampal gene expression of some chromatin-modifying enzyme genes, such as Dnmt3b. Hdac1. Hdac2. Sirt2, and Sirt6. Subsequently, we assessed the effects of EE on neuroprotection-related transcription factors, such as the Nuclear regulatory factor 2 (Nrf2)-Antioxidant Response Element pathway and Nuclear Factor kappa Beta (NF-κB), which play critical roles in inflammation. We found that EE produces an increased expression of antioxidant genes, such as Hmox1. Aox1, and Cox2, and reduced the expression of inflammatory genes such as IL-6 and Cxcl10, all of this within the epigenetic context modified by EE. In conclusion, EE prevents epigenetic changes that promote or drive oxidative stress and inflammaging.
Collapse
Affiliation(s)
- Christian Griñan-Ferré
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona Barcelona, Spain
| | - Dolors Puigoriol-Illamola
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona Barcelona, Spain
| | - Verónica Palomera-Ávalos
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona Barcelona, Spain
| | - David Pérez-Cáceres
- Animal Experimentation Unit, Faculty of Pharmacy, University of Barcelona Barcelona, Spain
| | - Júlia Companys-Alemany
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona Barcelona, Spain
| | - Antonio Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona Barcelona, Spain
| | - Daniel Ortuño-Sahagún
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara Guadalajara, Mexico
| | - M Teresa Rodrigo
- Animal Experimentation Unit, Faculty of Pharmacy, University of Barcelona Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona Barcelona, Spain
| |
Collapse
|