1
|
Abedini A, Landry DA, Macaulay AD, Vaishnav H, Parbhakar A, Ibrahim D, Salehi R, Maranda V, Macdonald E, Vanderhyden BC. SWI/SNF chromatin remodeling subunit Smarca4/BRG1 is essential for female fertility†. Biol Reprod 2023; 108:279-291. [PMID: 36440965 PMCID: PMC9930400 DOI: 10.1093/biolre/ioac209] [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: 11/10/2022] [Revised: 07/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Mammalian folliculogenesis is a complex process that involves the regulation of chromatin structure for gene expression and oocyte meiotic resumption. The SWI/SNF complex is a chromatin remodeler using either Brahma-regulated gene 1 (BRG1) or BRM (encoded by Smarca4 and Smarca2, respectively) as its catalytic subunit. SMARCA4 loss of expression is associated with a rare type of ovarian cancer; however, its function during folliculogenesis remains poorly understood. In this study, we describe the phenotype of BRG1 mutant mice to better understand its role in female fertility. Although no tumor emerged from BRG1 mutant mice, conditional depletion of Brg1 in the granulosa cells (GCs) of Brg1fl/fl;Amhr2-Cre mice caused sterility, whereas conditional depletion of Brg1 in the oocytes of Brg1fl/fl;Gdf9-Cre mice resulted in subfertility. Recovery of cumulus-oocyte complexes after natural mating or superovulation showed no significant difference in the Brg1fl/fl;Amhr2-Cre mutant mice and significantly fewer oocytes in the Brg1fl/fl;Gdf9-Cre mutant mice compared with controls, which may account for the subfertility. Interestingly, the evaluation of oocyte developmental competence by in vitro culture of retrieved two-cell embryos indicated that oocytes originating from the Brg1fl/fl;Amhr2-Cre mice did not reach the blastocyst stage and had higher rates of mitotic defects, including micronuclei. Together, these results indicate that BRG1 plays an important role in female fertility by regulating granulosa and oocyte functions during follicle growth and is needed for the acquisition of oocyte developmental competence.
Collapse
Affiliation(s)
- Atefeh Abedini
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - David A Landry
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Angus D Macaulay
- Chronic Diseases Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Het Vaishnav
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Ashna Parbhakar
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Dalia Ibrahim
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Reza Salehi
- Chronic Diseases Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Vincent Maranda
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Elizabeth Macdonald
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Barbara C Vanderhyden
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| |
Collapse
|
2
|
Zhou J, Yue S, Du J, Xue B, Wang L, Peng Q, Zou H, Hu R, Jiang Y, Wang Z, Xue B. Integration of transcriptomic and metabolomic analysis of the mechanism of dietary N-carbamoylglutamate in promoting follicle development in yaks. Front Vet Sci 2022; 9:946893. [PMID: 36105003 PMCID: PMC9464987 DOI: 10.3389/fvets.2022.946893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Yak is the main livestock in the highlands of China. The low reproductive rate of yaks is a serious constraint on their production and utility. N-carbamylglutamate (NCG) can increase arginine synthesis in mammals and has been shown to improve reproductive performance. Twelve multiparous and simutaneous anoestrous female yaks were randomly divided into two groups, one of which was fed the basal diet (Control, n = 6), and the other was fed the basal diet supplemented with NCG at 6 g/day/yak (NCG, n = 6). All yaks were slaughtered on the 32nd day (the time predicted for the selection of the last wave of dominant follicles), and their ovarian tissues were collected and follicles were classified. NCG supplementation increased the number of large ovarian follicles (diameter > 10 mm), as well as caused significant changes in the transcriptional and metabolic levels in yak ovaries which due to the differential expression of 889 genes and 94 metabolites. Integrated analysis of the transcriptomics and metabolomics data revealed that the differentially expressed genes and differential metabolites were primarily involved in the process of energy metabolism, amino acid metabolic pathways, carbohydrate metabolic pathways, and lipid metabolic pathways. The highlighted changes were associated with amino acid synthesis and metabolism, ovarian steroid hormone synthesis, the pentose phosphate pathway, and the tricarboxylic acid cycle, suggesting that NCG supplementation may promote estrogen synthesis and help regulate follicular development by altering the pathways associated with glucose catabolism. The results present important clues for understanding the mechanisms by which NCG supplementation promotes follicular development in yaks. The findings of this study provide a basis for the development and application of NCG in optimizing animal reproduction, including yak reproductive performance, which may help optimize livestock management and uplift the pastoral economy.
Collapse
Affiliation(s)
- Jia Zhou
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Shuangming Yue
- Department of Bioengineering, Sichuan Water Conservancy College, Chengdu, China
| | - Jingjing Du
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Benchu Xue
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Lizhi Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Quanhui Peng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Huawei Zou
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Rui Hu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Yahui Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhisheng Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Bai Xue
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Bai Xue
| |
Collapse
|
3
|
Xiao H, Xu Z, Zhu X, Wang J, Zheng Q, Zhang Q, Xu C, Tao W, Wang D. Cortisol safeguards oogenesis by promoting follicular cell survival. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1563-1577. [PMID: 35167018 DOI: 10.1007/s11427-021-2051-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
The role of glucocorticoids in oogenesis remains to be elucidated. cyp11c1 encodes the key enzyme involved in the synthesis of cortisol, the major glucocorticoid in teleosts. In our previous study, we mutated cyp11c1 in tilapia and analyzed its role in spermatogenesis. In this study, we analyzed its role in oogenesis. cyp11c1+/- XX tilapia showed normal ovarian morphology but poor egg quality, as indicated by the mortality of embryos before 3 d post fertilization, which could be partially rescued by the supplement of exogenous cortisol to the mother fish. Transcriptome analyses revealed reduced expression of maternal genes in the eggs of the cyp11c1+/- XX fish. The cyp11c1-/- females showed impaired vitellogenesis and arrested oogenesis due to significantly decreased serum cortisol. Further analyses revealed decreased serum E2 level and expression of amh, an important regulator of follicular cell development, and increased follicular cell apoptosis in the ovaries of cyp11c1-/- XX fish, which could be rescued by supplement of either exogenous cortisol or E2. Luciferase assays revealed a direct regulation of cortisol and E2 on amh transcription via GRs or ESRs. Taken together, our results demonstrate that cortisol safeguards oogenesis by promoting follicular cell survival probably via Amh signaling.
Collapse
Affiliation(s)
- Hesheng Xiao
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Zhen Xu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Xi Zhu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Jingrong Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Qiaoyuan Zheng
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Qingqing Zhang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Chunmei Xu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Wenjing Tao
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
4
|
Mahmoud AA, Elfiky AM, Abo-Zeid FS. The anti-androgenic effect of quercetin on hyperandrogenism and ovarian dysfunction induced in a dehydroepiandrosterone rat model of polycystic ovary syndrome. Steroids 2022; 177:108936. [PMID: 34752810 DOI: 10.1016/j.steroids.2021.108936] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/15/2021] [Accepted: 10/29/2021] [Indexed: 12/21/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a multi-factorial endocrine disorder associated with hyperandrogenism. Dehydroepiandrosterone (DHEA) administration to prepubertal rats stimulates androgen biosynthesis and generation of the PCOS model. The present study aimed to evaluate the anti-androgenic effects of quercetin (Q) in comparison with metformin (MET) on hyperandrogenism and ovarian dysfunction in a DHEA-induced PCOS rat model. After induction of PCOS, female rats were allocated into six groups with 7 rats in each group: normal control; PCOS (DHEA), MET (25 mg/kg, oral administration), Q (25 mg/kg, oral administration), DHEA + MET (25 mg/kg, oral administration), and DHEA + Q (25 mg/kg, oral administration) for 28 days. MET and Q individually reduced body weight, serum free testosterone (T) and luteinizing hormone (LH), and LH/follicle-stimulating hormone (FSH) ratio in the PCOS rats. Both treatments elevated estradiol (E2) level, ovarian aromatase protein content, and E2/free T ratio in the PCOS rats. Additionally, MET and Q increased preantral, antral, and preovulatory follicles and corpora lutea counts, while both treatments decreased atretic follicle count and eliminated the formation of cysts in the PCOS rats. MET and Q reduced ovarian Bax and elevated Bcl-2 protein abundance in the PCOS rats. Our study revealed that Q is as effective as MET in reducing hyperandrogenism via decreasing free T level and improving hypothalamic-pituitary-ovarian axis function. The results suggest that MET and Q may enhance E2 concentration, ovarian aromatase protein content, folliculogenesis, and decrease atresia via attenuation of hyperandrogenism in PCOS rats.
Collapse
Affiliation(s)
- Asmaa A Mahmoud
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Asmaa M Elfiky
- Environmental and Occupational Medicine Department, Environmental Research Division, National Research Center, Cairo, Egypt
| | - Faten S Abo-Zeid
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
5
|
Hughes CHK, Murphy BD. Nuclear receptors: Key regulators of somatic cell functions in the ovulatory process. Mol Aspects Med 2020; 78:100937. [PMID: 33288229 DOI: 10.1016/j.mam.2020.100937] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/30/2022]
Abstract
The development of the ovarian follicle to its culmination by ovulation is an essential element of fertility. The final stages of ovarian follicular growth are characterized by granulosa cell proliferation and differentiation, and steroid synthesis under the influence of follicle-stimulating hormone (FSH). The result is a population of granulosa cells poised to respond to the ovulatory surge of luteinizing hormone (LH). Members of the nuclear receptor superfamily of transcription factors play indispensable roles in the regulation of these events. The key regulators of the final stages of follicular growth that precede ovulation from this family include the estrogen receptor beta (ESR2) and the androgen receptor (AR), with additional roles for others, including steroidogenic factor-1 (SF-1) and liver receptor homolog-1 (LRH-1). Following the LH surge, the mural and cumulus granulosa cells undergo rapid changes that result in expansion of the cumulus layer, and a shift in ovarian steroid hormone biosynthesis from estradiol to progesterone production. The nuclear receptor best associated with these events is LRH-1. Inadequate cumulus expansion is also observed in the absence of AR and ESR2, but not the progesterone receptor (PGR). The terminal stages of ovulation are regulated by PGR, which increases the abundance of the proteases that are directly responsible for rupture. It further regulates the prostaglandins and cytokines associated with the inflammatory-like characteristics of ovulation. LRH-1 regulates PGR, and is also a key regulator of steroidogenesis, cellular proliferation, and cellular migration, and cytoskeletal remodeling. In summary, nuclear receptors are among the panoply of transcriptional regulators with roles in ovulation, and several are necessary for normal ovarian function.
Collapse
Affiliation(s)
- Camilla H K Hughes
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Qc, J2S 2M2, Canada
| | - Bruce D Murphy
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Qc, J2S 2M2, Canada.
| |
Collapse
|
6
|
Lin E, Li Z, Huang Y, Ru G, He P. High Dosages of Equine Chorionic Gonadotropin Exert Adverse Effects on the Developmental Competence of IVF-Derived Mouse Embryos and Cause Oxidative Stress-Induced Aneuploidy. Front Cell Dev Biol 2020; 8:609290. [PMID: 33634101 PMCID: PMC7900142 DOI: 10.3389/fcell.2020.609290] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/21/2020] [Indexed: 02/05/2023] Open
Abstract
Gonadotropins play vital roles in the regulation of female reproductive ability and fertility. Our study aimed to determine the effects of superovulation induced by increasing doses of equine chorionic gonadotropin [eCG; also referred to as pregnant mare serum gonadotropin (PMSG)] on the developmental competence of mouse embryos and on aneuploidy formation during in vitro fertilization (IVF). eCG dose-dependently enhanced the oocyte yield from each mouse. Administration of 15 IU eCG significantly reduced the fertilization rate and the formation of four-cell embryos and blastocysts and increased the risk of chromosome aneuploidy. The IVF-derived blastocysts in the 15 IU eCG treatment group had the fewest total cells, inner cell mass (ICM) cells and trophectoderm (TE) cells. Moreover, more blastocysts and fewer apoptotic cells were observed in the 0, 5, and 10 IU eCG treatment groups than in the 15 IU eCG treatment group. We also investigated reactive oxygen species (ROS) levels and variations in several variables: mitochondrial membrane potential (MMP); active mitochondria; mitochondrial superoxide production; adenosine triphosphate (ATP) content; spindle structures; chromosome karyotypes; microfilament distribution; and the expression of Aurora B [an important component of the chromosomal passenger complex (CPC)], the spindle assembly checkpoint (SAC) protein mitotic arrest deficient 2 like 1 (MAD2L1), and the DNA damage response (DDR) protein γH2AX. Injection of 15 IU eCG increased ROS levels, rapidly reduced MMP, increased active mitochondria numbers and mitochondrial superoxide production, reduced ATP content, increased abnormal spindle formation rates, and induced abnormalities in chromosome number and microfilament distribution, suggesting that a high dose of eCG might alter developmental competence and exert negative effects on IVF-obtained mouse embryos. Additionally, the appearance of γH2AX and the significantly increased expression of Aurora B and MAD2L1 suggested that administration of relatively high doses of eCG caused Aurora B-mediated SAC activation triggered by ROS-induced DNA damage in early mouse IVF-derived embryos for self-correction of aneuploidy formation. These findings improve our understanding of the application of gonadotropins and provide a theoretical basis for gonadotropin treatment.
Collapse
|
7
|
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
|
8
|
The Effect of Steroid Hormones on Ovarian Follicle Development. VITAMINS AND HORMONES 2018; 107:155-175. [DOI: 10.1016/bs.vh.2018.01.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
9
|
Meinsohn MC, Morin F, Bertolin K, Duggavathi R, Schoonjans K, Murphy BD. The Orphan Nuclear Receptor Liver Homolog Receptor-1 (Nr5a2) Regulates Ovarian Granulosa Cell Proliferation. J Endocr Soc 2017; 2:24-41. [PMID: 29379893 PMCID: PMC5779114 DOI: 10.1210/js.2017-00329] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/22/2017] [Indexed: 12/29/2022] Open
Abstract
In mouse ovaries, liver receptor homolog-1 [nuclear receptor subfamily 5, group A,
member 2 (Nr5a2)] expression is restricted to granulosa cells. Mice with Nr5a2
depletion in this cell population fail to ovulate. To determine whether Nr5a2 is
essential for granulosa cell proliferation during follicular maturation, we generated
granulosa-specific conditional knockout mice (genotype Nr5a2 floxed Cre-recombinase
driven by the anti-Müllerian type II receptor, hereafter cKO) with Nr5a2
depletion from primary follicles forward. Proliferation in cKO granulosa cells was
substantially reduced relative to control (CON) counterparts, as assessed by
bromodeoxyuridine incorporation, proliferative cell nuclear antigen expression, and
fluorescent-activated cell sorting. Microarray analysis revealed >2000
differentially regulated transcripts between cKO and CON granulosa cells. Major gene
ontology pathways disrupted were proliferation, steroid biosynthesis, female gamete
formation, and ovulatory cycle. Transcripts for key cell-cycle genes, including
Ccnd1, Ccnd2, Ccne1,
Ccne2, E2f1, and E2f2, were in
reduced abundance. Transcripts from other cell-cycle-related factors, including
Cdh2, Plagl1, Cdkn1a,
Prkar2b, Gstm1, Cdk7, and
Pts, were overexpressed. Although the follicle-stimulating
hormone and estrogen receptors were overexpressed in the cKO animals, in
vivo treatment with estradiol-17β failed to
rescue decreased proliferation. In vitro inactivation of Nr5a2 using
the ML180 reverse agonist similarly decreased cell-cycle-related gene transcripts and
downstream targets, as in cKO mice. Pharmacological inhibition of
β-catenin, an Nr5a2 cofactor, decreased cyclin gene
transcripts and downstream targets. Terminal deoxynucleotidyltransferase-mediated
deoxyuridine triphosphate nick end labeling immunofluorescence and quantitative
polymerase chain reaction of pro/antiapoptotic and autophagic markers showed no
differences between cKO and CON granulosa cells. Thus, Nr5a2 is essential for
granulosa cell proliferation, but its depletion does not alter the frequency of
apoptosis nor autophagy.
Collapse
Affiliation(s)
- Marie-Charlotte Meinsohn
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Quebec J2S 2M2, Canada
| | - Fanny Morin
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Quebec J2S 2M2, Canada
| | - Kalyne Bertolin
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Quebec J2S 2M2, Canada
| | - Raj Duggavathi
- Department of Animal Science, McGill University, Ste-Anne de Bellevue, Qubec H9X 3V9, Canada
| | - Kristina Schoonjans
- Laboratory of Metabolic Signaling, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Bruce D Murphy
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Quebec J2S 2M2, Canada
| |
Collapse
|
10
|
Gámez-Del-Estal MM, Contreras I, Prieto-Pérez R, Ruiz-Rubio M. Epigenetic effect of testosterone in the behavior of C. elegans. A clue to explain androgen-dependent autistic traits? Front Cell Neurosci 2014; 8:69. [PMID: 24624060 PMCID: PMC3940884 DOI: 10.3389/fncel.2014.00069] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/17/2014] [Indexed: 12/04/2022] Open
Abstract
Current research indicates that the causes of autism spectrum disorders (ASDs) are multifactorial and include both genetic and environmental factors. To date, several works have associated ASDs with mutations in genes that encode proteins involved in neuronal synapses; however other factors and the way they can interact with the development of the nervous system remain largely unknown. Some studies have established a direct relationship between risk for ASDs and the exposure of the fetus to high testosterone levels during the prenatal stage. In this work, in order to explain possible mechanisms by which this androgenic hormone may interact with the nervous system, C. elegans was used as an experimental model. We observed that testosterone was able to alter the behavioral pattern of the worm, including the gentle touch response and the pharyngeal pumping rate. This impairment of the behavior was abolished using specific RNAi against genes orthologous to the human androgen receptor gene. The effect of testosterone was eliminated in the nhr-69 (ok1926) deficient mutant, a putative ortholog of human AR gene, suggesting that this gene encodes a receptor able to interact with the hormone. On the other hand the testosterone effect remained in the gentle touch response during four generations in the absence of the hormone, indicating that some epigenetic mechanisms could be involved. Sodium butyrate, a histone deacetylase inhibitor, was able to abolish the effect of testosterone. In addition, the lasting effect of testosterone was eliminated after the dauer stage. These results suggest that testosterone may impair the nervous system function generating transgenerational epigenetic marks in the genome. This work may provide new paradigms for understanding biological mechanisms involved in ASDs traits.
Collapse
Affiliation(s)
- M Mar Gámez-Del-Estal
- Departamento de Genética, Universidad de Córdoba, Hospital Universitario Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba Córdoba, Spain
| | - Israel Contreras
- Departamento de Genética, Universidad de Córdoba, Hospital Universitario Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba Córdoba, Spain
| | - Rocío Prieto-Pérez
- Departamento de Genética, Universidad de Córdoba, Hospital Universitario Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba Córdoba, Spain
| | - Manuel Ruiz-Rubio
- Departamento de Genética, Universidad de Córdoba, Hospital Universitario Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba Córdoba, Spain
| |
Collapse
|
11
|
Epigenetics of estrogen receptor signaling: role in hormonal cancer progression and therapy. Cancers (Basel) 2013; 3:1691-707. [PMID: 21814622 PMCID: PMC3147309 DOI: 10.3390/cancers3021691] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Estrogen receptor (ERα) signaling plays a key role in hormonal cancer progression. ERα is a ligand-dependent transcription factor that modulates gene transcription via recruitment to the target gene chromatin. Emerging evidence suggests that ERα signaling has the potential to contribute to epigenetic changes. Estrogen stimulation is shown to induce several histone modifications at the ERα target gene promoters including acetylation, phosphorylation and methylation via dynamic interactions with histone modifying enzymes. Deregulation of enzymes involved in the ERα-mediated epigenetic pathway could play a vital role in ERα driven neoplastic processes. Unlike genetic alterations, epigenetic changes are reversible, and hence offer novel therapeutic opportunities to reverse ERα driven epigenetic changes. In this review, we summarize current knowledge on mechanisms by which ERα signaling potentiates epigenetic changes in cancer cells via histone modifications.
Collapse
|
12
|
Fu S, Li Y, Huang J, Liu T, Hong Z, Chen A, Bast RC, Kavanagh JJ, Gershenson DM, Sood AK, Hu W. Aurora kinase inhibitor VE 465 synergistically enhances cytotoxicity of carboplatin in ovarian cancer cells through induction of apoptosis and downregulation of histone 3. Cancer Biol Ther 2012; 13:1034-41. [PMID: 22895067 DOI: 10.4161/cbt.21045] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Aurora kinases are essential for regulation of chromosome segregation and cytokinesis during mitosis and play a role in growth and progression of human tumors, including ovarian cancer. Aurora A and Aurora B are frequently overexpressed in high-grade and low-grade ovarian cancers. Targeting Aurora kinases has great potential for improving the efficacy of chemotherapies of ovarian cancer. In this study, we investigated whether the Aurora kinase inhibitor, VE 465, can enhance the anti-tumor activity of carboplatin in human ovarian cancer cells. The antitumor activity of VE 465 was tested by MTT proliferative assay in multiple established human epithelial ovarian cancer cell lines of varying p53 status. VE 465 and carboplatin had a synergistic effect on cell viability in both platinum-sensitive and -resistant ovarian cancers. The growth-inhibitory effect was accompanied by reduction in expression of histone 3 and an increase in apoptosis. We conclude that VE 465 enhances the efficacy of carboplatin agents in ovarian carcinoma.
Collapse
Affiliation(s)
- Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Bapat SA. Modulation of gene expression in ovarian cancer by active and repressive histone marks. Epigenomics 2012; 2:39-51. [PMID: 22122747 DOI: 10.2217/epi.09.38] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
DNA methylation and histone modifications often function concomitantly to drive an aberrant program of gene expression in most cancers. Consequently, they have also been identified as being associated with ovarian cancer. However, several basic issues remain unclear - are these marks established early during normal ovarian functioning, or at a preneoplastic stage, or through a gradual accumulation, or do they arise de novo during transformation? Such issues have been difficult to address in ovarian cancer wherein preneoplastic lesions and progression models have not yet been established and drug-refractive disease progression is rapid and aggressive. The review presents an overview of the known involvement of histone modifications in various cellular states that might contribute to our understanding of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Sharmila A Bapat
- National Centre for Cell Science, NCCS complex, Pune University Campus, Ganeshkhind, Pune, India.
| |
Collapse
|
14
|
Kwintkiewicz J, Padilla-Banks E, Jefferson WN, Jacobs IM, Wade PA, Williams CJ. Metastasis-associated protein 3 (MTA3) regulates G2/M progression in proliferating mouse granulosa cells. Biol Reprod 2012; 86:1-8. [PMID: 22075476 PMCID: PMC3316264 DOI: 10.1095/biolreprod.111.096032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 09/18/2011] [Accepted: 10/25/2011] [Indexed: 01/10/2023] Open
Abstract
Metastasis-associated protein 3 (MTA3) is a constituent of the Mi-2/nucleosome remodeling and deacetylase (NuRD) protein complex that regulates gene expression by altering chromatin structure and can facilitate cohesin loading onto DNA. The biological function of MTA3 within the NuRD complex is unknown. Herein, we show that MTA3 was expressed highly in granulosa cell nuclei of all ovarian follicle stages and at lower levels in corpora lutea. We tested the hypothesis that MTA3-NuRD complex function is required for granulosa cell proliferation. In the ovary, MTA3 interacted with NuRD proteins CHD4 and HDAC1 and the core cohesin complex protein RAD21. In cultured mouse primary granulosa cells, depletion of endogenous MTA3 using RNA interference slowed cell proliferation; this effect was rescued by coexpression of exogenous MTA3. Slowing of cell proliferation correlated with a significant decrease in cyclin B1 and cyclin B2 expression. Granulosa cell populations lacking MTA3 contained a significantly higher percentage of cells in G2/M phase and a lower percentage in S phase compared with control cells. Furthermore, MTA3 depletion slowed entry into M phase as indicated by reduced phosphorylation of histone H3 at serine 10. These findings provide the first evidence to date that MTA3 interacts with NuRD and cohesin complex proteins in the ovary in vivo and regulates G2/M progression in proliferating granulosa cells.
Collapse
Affiliation(s)
- Jakub Kwintkiewicz
- Reproductive Medicine Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Elizabeth Padilla-Banks
- Reproductive Medicine Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Wendy N. Jefferson
- Reproductive Medicine Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Ilana M. Jacobs
- Reproductive Medicine Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Paul A. Wade
- Eukaryotic Transcriptional Regulation Group, Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Carmen J. Williams
- Eukaryotic Transcriptional Regulation Group, Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
15
|
Current advances in epigenetic modification and alteration during mammalian ovarian folliculogenesis. J Genet Genomics 2012; 39:111-23. [PMID: 22464470 DOI: 10.1016/j.jgg.2012.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 01/07/2012] [Accepted: 02/10/2012] [Indexed: 11/23/2022]
Abstract
During the growth and development of mammalian ovarian follicles, the activation and deactivation of mass genes are under the synergistic control of diverse modifiers through genetic and epigenetic events. Many factors regulate gene activity and functions through epigenetic modification without altering the DNA sequence, and the common mechanisms may include but are not limited to: DNA methylation, histone modifications (e.g., acetylation, deacetylation, phosphorylation, methylation, and ubiquitination), and RNA-associated silencing of gene expression by noncoding RNA. Over the past decade, substantial progress has been achieved in studies involving the epigenetic alterations during mammalian germ cell development. A number of candidate regulatory factors have been identified. This review focuses on the current available information of epigenetic alterations (e.g., DNA methylation, histone modification, noncoding-RNA-mediated regulation) during mammalian folliculogenesis and recounts when and how epigenetic patterns are differentially established, maintained, or altered in this process. Based on different types of epigenetic regulation, our review follows the temporal progression of events during ovarian folliculogenesis and describes the epigenetic changes and their contributions to germ cell-specific functions at each stage (i.e., primordial folliculogenesis (follicle formation), follicle maturation, and follicular atresia).
Collapse
|
16
|
Monga R, Ghai S, Datta TK, Singh D. Tissue-specific promoter methylation and histone modification regulate CYP19 gene expression during folliculogenesis and luteinization in buffalo ovary. Gen Comp Endocrinol 2011; 173:205-15. [PMID: 21663742 DOI: 10.1016/j.ygcen.2011.05.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 05/16/2011] [Accepted: 05/24/2011] [Indexed: 01/08/2023]
Abstract
Aromatase, the key enzyme of estrogen biosynthesis, is encoded by the CYP19 gene. The expression of CYP19 gene is regulated in species- and tissue-specific manner by alternate use of different promoters. We have previously, cloned and characterized the tissue-specific promoter and tissue-specific transcripts in preovulatory (granulosa cells) and postovulatory (corpus luteum) structure of buffalo ovary. The present study was aimed to understand if epigenetic gene regulation through DNA methylation and histone modifications is involved in tissue-specific CYP19 gene regulation during folliculogenesis and luteinization in buffalo ovary. Methylation analysis of five CpG dinucleotides of ovary specific proximal promoter II showed hypo-methylation in large follicle while hyper-methylation in corpus luteum. However, PI.1, the exclusive promoter responsible for residual CYP19 gene expression in corpus luteum, was found to be hypermethylated. Analysis of histone modifications using ChIP assay revealed that the distal promoter (PI.1) of CYP19 gene is ~40-fold more enriched with acetylated Histone H3 in corpus luteum than in the large follicle. This indicates that PI.1 chromatin was more accessible for transcription in corpus luteum as compared to large follicles. The chromatin accessibility for the proximal promoter (PII) in the preovulatory stage tends to be higher than the luteal tissue. However, the difference was not found to be significant. In vitro experiments showed the similar results. In conclusion, results of the present study suggests that tissue-specific methylation status of PII and chromatin remodeling through histone modifications of PI.1, coincides with the changes in expression of CYP19 gene and thus are the regulatory mechanism controlling its tissue-specific expression and promoter activity during folliculogenesis and luteinization.
Collapse
Affiliation(s)
- Rachna Monga
- Molecular Endocrinology Laboratory, Animal Biochemistry Division, National Dairy Research Institute, Karnal, Haryana, India
| | | | | | | |
Collapse
|
17
|
Abstract
Dynamic changes in chromatin structure and gene expression occur during follicular and oocyte growth. Epigenetic mechanisms regulate these changes through biochemical reactions that modify the nucleosome structure, and consequently affect transcription. Chromatin remodellers that alter DNA-histone interactions can influence transcriptional activity by facilitating or repressing DNA access. The SWItch/Sucrose NonFermentable (SWI/SNF) complex represents an important chromatin remodelling family, which comprises many protein subunits including the BRG1 (brahma-related gene 1). Our aim in this study was to analyse BRG1 expression patterns in different stages of follicular development. Ovaries (n = 10) were collected from prepubertal gilts and then rinsed in phosphate-buffered saline (PBS). Ovarian fragments of 8 × 8 × 8 mm were cut and placed into a 4% paraformaldehyde solution. For immunofluorescence analysis, samples were incubated with primary antibodies: polyclonal rabbit anti-BRG1 (1/200) or control rabbit IgG at the same concentration, overnight at 4°C. Primary antibodies were detected using Alexa Fluor 594-anti-rabbit 1/1000 diluted secondary antibody. Cells were counterstained with 4',6-diamidino-2-phenylindole (DAPI). Positive fluorescence signal for BRG1 was detected in all analysed samples. In primary follicles, the protein was detected only in the oocyte nucleus. However, in growing follicles, BRG1 was identified in granulosa and theca cells in a well defined pattern, according to the proximity of the cells from the oocyte. These results suggest an important role for BRG1 in the regulation of follicular growth, probably modulating granulosa and theca cell proliferation, as well as oocyte growth and maturation.
Collapse
|
18
|
Tardáguila M, González-Gugel E, Sánchez-Pacheco A. Aurora kinase B activity is modulated by thyroid hormone during transcriptional activation of pituitary genes. Mol Endocrinol 2011; 25:385-93. [PMID: 21239609 DOI: 10.1210/me.2010-0446] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Covalent histone modifications clearly play an essential role in ligand-dependent transcriptional regulation by nuclear receptors. One of the predominant mechanisms used by nuclear receptors to activate or repress target-gene transcription is the recruitment of coregulatory factors capable of covalently modify the amino terminal ends of histones. Here we show that the thyroid hormone (T3) produces a rapid increase in histone H3Ser10 phosphorylation (H3Ser10ph) concomitant to the rapid displacement of the heterochromatin protein 1β (HP1β) to the nuclear periphery. Moreover, we found that T3-mediated pituitary gene transcription is associated with an increase in H3Ser10ph. Interestingly, the Aurora kinase B inhibitor ZM443979 abolishes the effect of T3 on H3Ser10ph, blocks HP1β delocalization, and significantly reduces ligand-dependent transactivation. Similar effects were shown when Aurora kinase B expression was abrogated in small interfering RNA assays. In an effort to understand the underlying mechanism by which T3 increases H3Ser10ph, we demonstrate that liganded thyroid hormone receptor directly interacts with Aurora kinase B, increasing its kinase activity. Moreover, using chromatin immunoprecipitation assays, we have shown that Aurora kinase B participates of a mechanism that displaces HP1β from promoter region, thus preparing the chromatin for the transcriptional activation of T3 regulated genes. Our findings reveal a novel role for Aurora kinase B during transcriptional initiation in GO/G1, apart from its well-known mitotic activity.
Collapse
Affiliation(s)
- Manuel Tardáguila
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Arturo Duperier 4, 28029 Madrid, Spain
| | | | | |
Collapse
|
19
|
Binelli M, Murphy BD. Coordinated regulation of follicle development by germ and somatic cells. Reprod Fertil Dev 2010; 22:1-12. [PMID: 20003840 DOI: 10.1071/rd09218] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The continuum of folliculogenesis begins in the fetal ovary with the differentiation of the oogonia and their isolation within the primordial follicles. Primordial follicle activation is an enigmatic process, whereby some follicles enter the growing pool to become primary follicles, thereby embarking on an irreversible progression towards ovulation or atresia. This process is under the coordinated regulation of factors from the oocyte itself, as well as from the somatic cells of the ovary, in particular the theca and granulosa cells, which are structural components of the follicle. These two influences provide the principal stimuli for the growth of the follicle to the late preantral or early antral stage of development. The endocrine effects of the gonadotrophins FSH and LH are essential to the continued progression of the follicle and most atresia can be attributed to the failure to receive or process the gonadotrophin signals. The peri-ovulatory state has received intensive investigation recently, demonstrating a coordinated role for gonadotrophins, steroids, epidermal growth factor family proteins and prostaglandins. Thus, a complex programme of coordinated interaction of governing elements from both germ and somatic cell sources is required for successful follicle development.
Collapse
Affiliation(s)
- Mario Binelli
- College of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, SP 13635-900, Brazil
| | | |
Collapse
|
20
|
Murray EK, Hien A, de Vries GJ, Forger NG. Epigenetic control of sexual differentiation of the bed nucleus of the stria terminalis. Endocrinology 2009; 150:4241-7. [PMID: 19497973 PMCID: PMC2736071 DOI: 10.1210/en.2009-0458] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The principal nucleus of the bed nucleus of the stria terminalis (BNSTp) is larger in volume and contains more cells in male than female mice. These sex differences depend on testosterone and arise from a higher rate of cell death during early postnatal life in females. There is a delay of several days between the testosterone surge at birth and sexually dimorphic cell death in the BNSTp, suggesting that epigenetic mechanisms may be involved. We tested the hypothesis that chromatin remodeling plays a role in sexual differentiation of the BNSTp by manipulating the balance between histone acetylation and deacetylation using a histone deacetylase inhibitor. In the first experiment, a single injection of valproic acid (VPA) on the day of birth increased acetylation of histone H3 in the brain 24 h later. Next, males, females, and females treated neonatally with testosterone were administered VPA or saline on postnatal d 1 and 2 and killed at 21 d of age. VPA treatment did not influence volume or cell number of the BNSTp in control females but significantly reduced both parameters in males and testosterone-treated females. As a result, the sex differences were eliminated. VPA did not affect volume or cell number in the suprachiasmatic nucleus or the anterodorsal nucleus of the thalamus, which also did not differ between males and females. These findings suggest that a disruption in histone deacetylation may lead to long-term alterations in gene expression that block the masculinizing actions of testosterone in the BNSTp.
Collapse
Affiliation(s)
- Elaine K Murray
- Department of Psychology and Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | | | | | | |
Collapse
|
21
|
Price DM, Kanyo R, Steinberg N, Chik CL, Ho AK. Nocturnal activation of aurora C in rat pineal gland: its role in the norepinephrine-induced phosphorylation of histone H3 and gene expression. Endocrinology 2009; 150:2334-41. [PMID: 19116339 DOI: 10.1210/en.2008-1507] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have shown previously that Ser10 phosphorylation of histone H3 occurs in rat pinealocytes after stimulation with norepinephrine (NE) and that histone modifications such as acetylation appear to play an important role in pineal gene transcription. Here we report the nocturnal phosphorylation of a Ser10 histone H3 kinase, Aurora C, in the rat pineal gland. The time profile of this phosphorylation parallels the increase in the level of phospho-Ser10 histone H3. Studies with cultured pinealocytes indicate that Aurora C phosphorylation is induced by NE and this induction can be blocked by cotreatment with propranolol or KT5720, a protein kinase A inhibitor. Moreover, only treatment with dibutyryl cAMP, but not other kinase activators, mimics the effect of NE on Aurora C phosphorylation. These results indicate that Aurora C is phosphorylated primarily by a beta-adrenergic/protein kinase A-mediated mechanism. Treatment with an Aurora C inhibitor reduces the NE-induced histone H3 phosphorylation and suppresses the NE-stimulated induction of arylalkylamine N-acetyltransferase (AA-NAT), the rhythm-controlling enzyme of melatonin synthesis, and melatonin production. The effects of Aurora C inhibitors on adrenergic-induced genes in rat pinealocytes are gene specific: inhibitory for Aa-nat and inducible cAMP repressor but stimulatory for c-fos. Together our results support a role for the NE-stimulated phosphorylation of Aurora C and the subsequent remodeling of chromatin in NE-stimulated Aa-nat transcription. This phenomenon suggests that activation of this mitotic kinase can be induced by extracellular signals to participate in the transcriptional induction of a subset of genes in the rat pineal gland.
Collapse
Affiliation(s)
- D M Price
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | | | | | | | | |
Collapse
|
22
|
Chiam K, Tilley WD, Butler LM, Bianco-Miotto T. The dynamic and static modification of the epigenome by hormones: a role in the developmental origin of hormone related cancers. Biochim Biophys Acta Rev Cancer 2009; 1795:104-9. [PMID: 19166905 DOI: 10.1016/j.bbcan.2008.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 12/14/2008] [Accepted: 12/21/2008] [Indexed: 12/19/2022]
Abstract
There are numerous diseases associated with abnormal hormonal regulation and these include cancers of the breast and prostate. There is substantial evidence that early hormonal perturbations (in utero or during early development) are associated with increased disease susceptibility later in life. These perturbations may arise from exposure to environmental agents or endocrine disruptors which mimic hormones and disrupt normal hormonal signaling. Epigenetic alterations have often been proposed as the underlying mechanism by which early hormonal perturbations may give rise to disease in adulthood. Currently, there is minimal evidence to support a direct link between early hormonal perturbations and epigenetic modifications; or between epigenetic alterations and subsequent onset of cancer. Given that epigenetic modifications may play an important role in hormone-dependent cancers, it is essential to better understand the relationship between the hormonal environment and epigenetic modifications in both normal and disease states. In this review, we highlight several important studies which support the hypothesis that: hormonal perturbations early in life may result in epigenetic changes that may modify hormone receptor function, thereby contributing to an increased risk of developing hormone-related cancers.
Collapse
Affiliation(s)
- Karen Chiam
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, Hanson Institute, PO Box 14 Rundle Mall, Adelaide, South Australia, 5000, Australia
| | | | | | | |
Collapse
|
23
|
Seneda MM, Godmann M, Murphy BD, Kimmins S, Bordignon V. Developmental regulation of histone H3 methylation at lysine 4 in the porcine ovary. Reproduction 2008; 135:829-38. [PMID: 18502896 DOI: 10.1530/rep-07-0448] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Follicular growth and oogenesis involve highly dynamic changes in morphogenesis, chromatin structure, and gene transcription. The tight coordination of these events leads to ovulation of a mature oocyte and formation of the luteal tissue necessary to regulate embryo implantation and development. This entire process is regulated by numerous endocrine and in situ mechanisms. The role of epigenetic mechanisms in folliculogenesis, such as the biochemical modification of the DNA packaging proteins, the histones, is not well understood. Our objective was to determine the cellular and follicular stage-specific patterns of histone H3 methylation at lysine 4 (K4) in porcine preovulatory follicles and during luteinization in pig ovaries. Ovary tissues were collected from slaughtered prepubertal and cyclic gilts at various stages of the estrous cycle, pregnancy, and from ovaries recovered from gonatropin-treated gilts at 0, 24, and 38 h post human chorionic gonadotropin (hCG) injection. Samples were fixed in 4% paraformaldehyde and processed for embedding in paraffin and sectioned using standard histological protocols. Immunofluorescent staining was performed on 3 microm thick sections. The immunostaining pattern of mono-, di-, and tri-methylated histone H3-K4 and lysine-specific demethylase 1 (LSD1, also known as KDM1 or AOF1) was assessed. Interestingly, H3-K4 mono-, di-, and tri-methylation in follicles of prepubertal gilts was specifically distributed and developmentally regulated. While granulosa cells of primary, secondary, and early antral follicles were negative for H3-K4 methylation those from large antral follicles showed a striking upregulation in the cells located in the proximity to the oocyte. Specifically, the cumulus oophorus displayed intense staining for H3-K4 methylation and signals were strongest in the granulosa cells in the inner two cell layers of the follicular wall. Although all oocytes from primary to large antral stage follicles were positive for H3-K4 mono-, di-, and tri-methylation, the patterns of distribution were altered through oocyte follicle development. H3-K4 methylation in granulosa cells was dramatically reduced as time to ovulation approached and was low to undetected at 38 h post hCG treatment. H3-K4 mono-, di-, and tri-methylation in large luteal cells increased as differentiation evolved but remained low in small luteal cells. Strikingly, LSD1 (KDM1) expression was found to be restricted to the corpus luteum. In summary, this study provides new information on histone H3-K4 methylation patterns in the oocyte and follicle during folliculogenesis, which suggests that these epigenetic markers serve an essential regulatory role during folliculogenesis.
Collapse
Affiliation(s)
- Marcelo M Seneda
- Departamento de Clínicas Veterinárias, Universidade Estadual de Londrina, Londrina, Paraná, 86051-990, Brasil
| | | | | | | | | |
Collapse
|
24
|
Chu S, Alexiadis M, Fuller PJ. Proteasome Inhibition by Bortezomib Decreases Proliferation and Increases Apoptosis in Ovarian Granulosa Cell Tumors. Reprod Sci 2008; 16:397-407. [DOI: 10.1177/1933719108327589] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Simon Chu
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| | - Maria Alexiadis
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| | - Peter J. Fuller
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia,
| |
Collapse
|
25
|
Nadler Y, Camp RL, Schwartz C, Rimm DL, Kluger HM, Kluger Y. Expression of Aurora A (but not Aurora B) is predictive of survival in breast cancer. Clin Cancer Res 2008; 14:4455-62. [PMID: 18628459 DOI: 10.1158/1078-0432.ccr-07-5268] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE The cell cycle mediators Aurora A and B are targets of drugs currently in clinical development. As with other targeted therapies in breast cancer, response to therapy might be associated with target expression in tumors. We therefore assessed expression of Aurora A and B in breast tumors and studied associations with clinical/pathologic variables. EXPERIMENTAL DESIGN Tissue microarrays containing primary specimens from 638 patients with 15-year follow-up were employed to assess expression of Aurora A and B using our automated quantitative analysis method; we used cytokeratin to define pixels as breast cancer (tumor mask) within the array spot and measured Aurora A and B expression within the mask using Cy5-conjugated antibodies. RESULTS Aurora A and B expression was variable in primary breast tumors. High Aurora A expression was strongly associated with decreased survival (P = 0.0005). On multivariable analysis, it remained an independent prognostic marker. High Aurora A expression was associated with high nuclear grade and high HER-2/neu and progesterone receptor expression. Aurora B expression was not associated with survival. CONCLUSIONS Aurora A expression defines a population of patients with decreased survival, whereas Aurora B expression does not, suggesting that Aurora A might be the preferred drug target in breast cancer. Aurora A expression in early-stage breast cancer may identify a subset of patients requiring more aggressive or pathway-targeted treatment. Prospective studies are needed to confirm the prognostic role of Aurora A as well as the predictive role of Aurora A expression in patients treated with Aurora A inhibitors.
Collapse
Affiliation(s)
- Yasmine Nadler
- Department of Medicine and Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
26
|
Lazzaro MA, Pépin D, Pescador N, Murphy BD, Vanderhyden BC, Picketts DJ. The imitation switch protein SNF2L regulates steroidogenic acute regulatory protein expression during terminal differentiation of ovarian granulosa cells. Mol Endocrinol 2006; 20:2406-17. [PMID: 16740656 DOI: 10.1210/me.2005-0213] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Luteinization is a complex process, stimulated by gonadotropins, that promotes ovulation and development of the corpus luteum through terminal differentiation of granulosa cells. The pronounced expression of the mammalian imitation switch (ISWI) genes, SNF2H and SNF2L, in adult ovaries prompted us to investigate the role of these chromatin remodeling proteins during follicular development and luteinization. SNF2H expression is highest during growth of preovulatory follicles and becomes less prevalent during luteinization. In contrast, both SNF2L transcript and SNF2L protein levels are rapidly increased in granulosa cells of the mouse ovary 8 h after human chorionic gonadotropin treatment, and continue to be expressed 36 h later within the functional corpus luteum. We demonstrate a physical interaction between SNF2L and the progesterone receptor A isoform, which regulates progesterone receptor-responsive genes required for ovulation. Moreover, chromatin immunoprecipitation demonstrated that, after gonadotropin stimulation, SNF2L is associated with the proximal promoter of the steroidogenic acute regulatory protein (StAR) gene, a classic marker of luteinization in granulosa cells. Interaction of SNF2L with the StAR promoter is required for StAR expression, because small interfering RNA knockdown of SNF2L prevents the activation of the StAR gene. Our results provide the first indication that ISWI chromatin remodeling proteins are responsive to the LH surge and that this response is required for the activation of the StAR gene and the overall development of a functional luteal cell.
Collapse
Affiliation(s)
- Maribeth A Lazzaro
- Molecular Medicine Program, Ottawa Health Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | | | | | | | | | | |
Collapse
|