1
|
Ulloa-Aguirre A, Zariñán T, Dias JA, Kumar TR, Bousfield GR. Biased signaling by human follicle-stimulating hormone variants. Pharmacol Ther 2025; 268:108821. [PMID: 39961417 DOI: 10.1016/j.pharmthera.2025.108821] [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: 10/15/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/23/2025]
Abstract
Follicle-stimulating hormone (FSH) or follitropin plays a fundamental role in several mammalian species, including humans. This gonadotropin is produced by the anterior pituitary gland and has as its main targets the granulosa cells of the ovary and the Sertoli cells of the testis. Structurally, FSH is composed of two non-convalently linked subunits, the α- and β-subunit, as well as highly heterogenous oligosaccharide structures, which play a key role in determining a number of physiological and biological features of the hormone. Glycosylation in FSH and the other members belonging to the glycoprotein hormone family, is essential for many functions of the gonadotropin, including subunit assembly and stability, secretion, circulatory half-life and biological activity. Carbohydrate heterogeneity in FSH comes in two forms, microheterogeneity, which results from variations in the carbohydrate structural complexity in those oligosaccharides attached to the α- or β-subunit of the hormone and macroheterogeneity, which results from the absence of carbohydrate chain at FSHβ Asn-glycosylation sites. A number of in vitro and in vivo studies have conclusively demonstrated differential, unique and even opposing effects provoked by variations in the carbohydrate structures of FSH, including circulatory survival, binding to and activation of its cognate receptor in the gonads, intracellular signaling, and activation/inhibition of a number of FSH-regulated genes essential for follicle development. Herein, we review the effects of the FSH oligosaccharides on several functions of FSH, and how variations in these structures have been shown to lead to functional selectivity of the hormone.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico..
| | - Teresa Zariñán
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - James A Dias
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, USA
| | - T Rajendra Kumar
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - George R Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, KS, USA
| |
Collapse
|
2
|
Southall J, Park S, Choi Y, Jeon H, Ko C, Jo M. Granulosa cell expression of Fos is critical for regulating ovulatory gene expressions in the mouse ovary. FASEB J 2025; 39:e70388. [PMID: 39945297 PMCID: PMC11922626 DOI: 10.1096/fj.202402867r] [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: 11/19/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 03/20/2025]
Abstract
A previous study showed that female Fos null mice fail to ovulate even when given gonadotropins, suggesting that ovarian expression of Fos is critical for successful ovulation. However, the expression of FOS and function of FOS have not been determined in the mouse ovary. FOS, a member of the Fos family (Fos, Fosb, Fosl1, and Fosl2), functions as a transcription factor by forming a heterodimer complex with a member of Jun family (Jun, Junb, and Jund). This study demonstrated rapid increases in Fos, along with other Fos and Jun family members, after hCG administration in the ovary of immature PMSG-primed mice and after the LH surge in naturally cycling animals. ChIP-seq analysis identified 1965 FOS-binding genes in granulosa cells collected at 3 h post-hCG, including Pgr, Ptgs2, Tnfiap6, and Edn2, genes known to be involved in the ovulatory process. When super-ovulation was induced, the number of oocytes released was significantly reduced in Esr2cre/+-driven granulosa cell-specific Fos knockout (gcFosKO) mice. This reduction was accompanied by lower expressions of Pgr, Ptgs2, Ptgs1, and Edn2 in preovulatory follicles of gcFosKO mice compared to those in control littermates. In addition, gcFosKO mice showed a trend toward a decreased average litter size. Together, the present study indicates that the preovulatory induction of Fos expression is crucial for increasing the expression of key ovulatory genes, yet the role of FOS may be partially substituted by other Fos and Jun family members induced in the preovulatory follicle in the gcFosKO mouse ovary.
Collapse
Affiliation(s)
- Jacqueline Southall
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Shawn Park
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Yohan Choi
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Hayce Jeon
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Chemyong Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
3
|
Walters K, Baldwin A, Liu Z, Larsen M, Mukherjee N, Kumar TR. Identification of FSH-regulated and estrous stage-specific transcriptional networks in mouse ovaries. Proc Natl Acad Sci U S A 2025; 122:e2411977122. [PMID: 39928863 PMCID: PMC11848299 DOI: 10.1073/pnas.2411977122] [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: 06/14/2024] [Accepted: 01/03/2025] [Indexed: 02/12/2025] Open
Abstract
Follicle-stimulating hormone (FSH) acts by binding to FSHRs expressed on ovarian granulosa cells and produces estradiol. FSH is essential for female fertility because mice lacking FSH (Fshb KO) are anestrous and infertile. Although several in vitro cell culture and ex vivo approaches combined with pharmacological hormone treatment were used to identify FSH-regulated genes, how FSH orchestrates ovarian gene networks in vivo has not been investigated. Whether FSH-regulated genes display estrous stage-specific expression changes has also not been studied. Here, we functionally rescued Fshb null mice with a gonadotrope-targeted HFSHB transgene and performed RNA-Seq analysis on ovarian RNAs obtained from FSH-intact (WT), FSH-deficient (Fshb KO), and FSH-rescue (HFSHB+ rescue) mice. By comparing WT vs. Fshb KO and Fshb KO vs. HFSHB+ rescue ovarian gene expression datasets, we identified FSH-responsive genes in vivo. Cross interrogation of these datasets further allowed us to identify several transcription factors (TFs) and RNA-binding proteins specific to FSH-regulated genes. In an independent set of experiments, we performed RNA-Seq analysis on ovarian RNAs from mice in diestrous (DE), proestrous (PE), and estrous (E) and identified estrous stage-specific ovarian gene expression patterns. Interestingly, many of the FSH-regulated TFs themselves were estrous-stage specifically expressed. We found that ESR2 and GATA6, two known FSH-responsive TFs, and their target genes are reciprocally regulated with distinct patterns of expression in estrous stages. Together, our in vivo models and RNA-Seq analyses identify FSH-regulated ovarian genes in specific estrous stages that are under transcriptional and posttranscriptional control.
Collapse
Affiliation(s)
- Kathryn Walters
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO80045
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Amber Baldwin
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO80045
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Zhenghui Liu
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Mark Larsen
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Neelanjan Mukherjee
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO80045
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - T. Rajendra Kumar
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| |
Collapse
|
4
|
Mączka K, Stasiak O, Przybysz P, Grymowicz M, Smolarczyk R. The Impact of the Endocrine and Immunological Function of Adipose Tissue on Reproduction in Women with Obesity. Int J Mol Sci 2024; 25:9391. [PMID: 39273337 PMCID: PMC11395521 DOI: 10.3390/ijms25179391] [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: 07/15/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Obesity, which leads to metabolic dysregulation and body function impairment, emerges as one of the pressing health challenges worldwide. Excessive body fat deposits comprise a dynamic and biologically active organ possessing its own endocrine function. One of the mechanisms underlying the pathophysiology of obesity is low-grade systemic inflammation mediated by pro-inflammatory factors such as free fatty acids, lipopolysaccharides, adipokines (including leptin, resistin and visfatin) and cytokines (TNF-α, IL-1β, Il-6), which are secreted by adipose tissue. Together with obesity-induced insulin resistance and hyperandrogenism, the exacerbated immune response has a negative impact on the hypothalamic-pituitary-gonadal axis at all levels and directly affects reproduction. In women, it results in disrupted ovarian function, irregular menstrual cycles and anovulation, contributing to infertility. This review focuses on the abnormal intracellular communication, altered gene expression and signaling pathways activated in obesity, underscoring its multifactorial character and consequences at a molecular level. Extensive presentation of the complex interplay between adipokines, cytokines, immune cells and neurons may serve as a foundation for future studies in search of potential sites for more targeted treatment of reproductive disorders related to obesity.
Collapse
Affiliation(s)
- Katarzyna Mączka
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Olga Stasiak
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
| | - Paulina Przybysz
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
| | - Monika Grymowicz
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
| | - Roman Smolarczyk
- Department of Gynecological Endocrinology, Medical University of Warsaw, 00-315 Warsaw, Poland
| |
Collapse
|
5
|
Esencan E, Beroukhim G, Seifer DB. Age-related changes in Folliculogenesis and potential modifiers to improve fertility outcomes - A narrative review. Reprod Biol Endocrinol 2022; 20:156. [PMID: 36397149 PMCID: PMC9670479 DOI: 10.1186/s12958-022-01033-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022] Open
Abstract
Reproductive aging is characterized by a decline in oocyte quantity and quality, which is directly associated with a decline in reproductive potential, as well as poorer reproductive success and obstetrical outcomes. As women delay childbearing, understanding the mechanisms of ovarian aging and follicular depletion have become increasingly more relevant. Age-related meiotic errors in oocytes are well established. In addition, it is also important to understand how intraovarian regulators change with aging and how certain treatments can mitigate the impact of aging. Individual studies have demonstrated that reproductive pathways involving antimullerian hormone (AMH), vascular endothelial growth factor (VEGF), neurotropins, insulin-like growth factor 1 (IGF1), and mitochondrial function are pivotal for healthy oocyte and cumulus cell development and are altered with increasing age. We provide a comprehensive review of these individual studies and explain how these factors change in oocytes, cumulus cells, and follicular fluid. We also summarize how modifiers of folliculogenesis, such as vitamin D, coenzyme Q, and dehydroepiandrosterone (DHEA) may be used to potentially overcome age-related changes and enhance fertility outcomes of aged follicles, as evidenced by human and rodent studies.
Collapse
Affiliation(s)
- Ecem Esencan
- Yale School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, New Haven, CT, USA.
| | - Gabriela Beroukhim
- Yale School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, New Haven, CT, USA
| | - David B Seifer
- Yale School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, New Haven, CT, USA
| |
Collapse
|
6
|
Jenabi M, Khodarahmi P, Tafvizi F, Bostanabad SZ. Evaluation of expression CXCL8 chemokine and its relationship with oocyte maturation and embryo quality in the intracytoplasmic sperm injection method. Mol Biol Rep 2022; 49:8413-8427. [PMID: 35781602 DOI: 10.1007/s11033-022-07660-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/30/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND The present study aimed to evaluate the expression of the chemokine CXCL8 in both mRNA and protein levels in the serum, follicular fluid (FF), and cumulus cells (CCs) and its relationship with oocyte maturation and embryo quality in women undergoing intracytoplasmic sperm injection (ICSI). METHODS A total of 87 women who underwent an ICSI cycle were evaluated in two groups, including the case group (female factor infertility) and the control group (fertile). In the serum, FF, and CCs, the protein and mRNA expression of CXCL8 were measured using immunosorbent assay and Real-Time PCR, respectively. The quality and quantity of the oocytes and embryos were assessed, and the relationship of protein and mRNA CXCL8 was evaluated with oocyte maturation and embryo quality. RESULTS The level of protein and mRNA of CXCL8 was significantly higher in the serum, FF, and CCs in the case group than in the control group. In the case group, the expression of mRNA and protein of CXCL8 had a significant increase in FF and CCs compared to serum; also, there was a CXCL8 protein significant increase in FF compared to CCs. The count of oocytes obtained, MII oocytes and the percentage of oocyte maturity significantly decreased in the case group. The expression of CXCL8 was inversely related to oocyte maturation, but no relationship was observed with embryo quality. CONCLUSIONS The elevated concentrations of CXCL8 in the serum and FF seem to be a predictor as a potential non-invasive biomarker for the oocyte maturation outcome in women with different causes of female factor infertility.
Collapse
Affiliation(s)
- Maryam Jenabi
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| | - Parvin Khodarahmi
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran.
| | - Farzaneh Tafvizi
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| | | |
Collapse
|
7
|
Bafor EE, Valencia JC, Young HA. Double Negative T Regulatory Cells: An Emerging Paradigm Shift in Reproductive Immune Tolerance? Front Immunol 2022; 13:886645. [PMID: 35844500 PMCID: PMC9283768 DOI: 10.3389/fimmu.2022.886645] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Immune regulation of female reproductive function plays a crucial role in fertility, as alterations in the relationship between immune and reproductive processes result in autoimmune subfertility or infertility. The breakdown of immune tolerance leads to ovulation dysfunction, implantation failure, and pregnancy loss. In this regard, immune cells with regulatory activities are essential to restore self-tolerance. Apart from regulatory T cells, double negative T regulatory cells (DNTregs) characterized by TCRαβ+/γδ+CD3+CD4–CD8– (and negative for natural killer cell markers) are emerging as effector cells capable of mediating immune tolerance in the female reproductive system. DNTregs are present in the female reproductive tract of humans and murine models. However, their full potential as immune regulators is evolving, and studies so far indicate that DNTregs exhibit features that can also maintain tolerance in the female reproductive microenvironment. This review describes recent progress on the presence, role and mechanisms of DNTregs in the female reproductive system immune regulation and tolerance. In addition, we address how DNTregs can potentially provide a paradigm shift from the known roles of conventional regulatory T cells and immune tolerance by maintaining and restoring balance in the reproductive microenvironment of female fertility.
Collapse
Affiliation(s)
- Enitome E Bafor
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Julio C Valencia
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Howard A Young
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| |
Collapse
|
8
|
Chen M, Dong F, Chen M, Shen Z, Wu H, Cen C, Cui X, Bao S, Gao F. PRMT5 regulates ovarian follicle development by facilitating Wt1 translation. eLife 2021; 10:68930. [PMID: 34448450 PMCID: PMC8483736 DOI: 10.7554/elife.68930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/26/2021] [Indexed: 01/20/2023] Open
Abstract
Protein arginine methyltransferase 5 (Prmt5) is the major type II enzyme responsible for symmetric dimethylation of arginine. Here, we found that PRMT5 was expressed at high level in ovarian granulosa cells of growing follicles. Inactivation of Prmt5 in granulosa cells resulted in aberrant follicle development and female infertility. In Prmt5-knockout mice, follicle development was arrested with disorganized granulosa cells in which WT1 expression was dramatically reduced and the expression of steroidogenesis-related genes was significantly increased. The premature differentiated granulosa cells were detached from oocytes and follicle structure was disrupted. Mechanism studies revealed that Wt1 expression was regulated by PRMT5 at the protein level. PRMT5 facilitated IRES-dependent translation of Wt1 mRNA by methylating HnRNPA1. Moreover, the upregulation of steroidogenic genes in Prmt5-deficient granulosa cells was repressed by Wt1 overexpression. These results demonstrate that PRMT5 participates in granulosa cell lineage maintenance by inducing Wt1 expression. Our study uncovers a new role of post-translational arginine methylation in granulosa cell differentiation and follicle development. Infertility in women can be caused by many factors, such as defects in the ovaries. An important part of the ovaries for fertility are internal structures called follicles, which house early forms of egg cells. A follicle grows and develops until the egg is finally released from the ovary into the fallopian tube, where the egg can then be fertilised. In the follicle, an egg is surrounded by other types of cells, such as granulosa cells. The egg and neighbouring cells must maintain healthy contacts with each other, otherwise the follicle can stop growing and developing, potentially causing infertility. The development of a follicle depends on an array of proteins. For example, the transcription factor WT1 controls protein levels by activating other genes and their proteins and is produced in high numbers by granulosa cells at the beginning of follicle development. Although WT1 levels dip towards the later stages of follicle development, insufficient levels can lead to defects. So far, it has been unclear how levels of WT1in granulose cells are regulated. Chen, Dong et al. studied mouse follicles to reveal more about the role of WT1 in follicle development. The researchers measured protein levels in mouse granulosa cells as the follicles developed, and discovered elevated levels of PRMT5, a protein needed for egg cells to form and survive in the follicles. Blocking granulosa cells from producing PRMT5 led to abnormal follicles and infertility in mice. Moreover, mice that had been engineered to lack PRMT5 developed abnormal follicles, where the egg and surrounding granulosa cells were not attached to each other, and the granulosa cells had low levels of WT1. Further experiments revealed that PRMT5 controlled WT1 levels by adding small molecules called methyl groups to another regulatory protein called HnRNPA1. The addition of methyl groups to genes or their proteins is an important modification that takes place in many processes within a cell. Chen, Dong et al. reveal that this activity also plays a key role in maintaining healthy follicle development in mice, and that PRMT5 is necessary for controlling WT1. Identifying all of the intricate mechanism involved in regulating follicle development is important for finding ways to combat infertility.
Collapse
Affiliation(s)
- Min Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Fangfang Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Min Chen
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zhiming Shen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Haowei Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Changhuo Cen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiuhong Cui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Shilai Bao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
9
|
Fang L, Yan Y, Wang S, Guo Y, Li Y, Jia Q, Han X, Liu B, Cheng JC, Sun YP. High ovarian GDF-8 levels contribute to elevated estradiol production in ovarian hyperstimulation syndrome by stimulating aromatase expression. Int J Biol Sci 2021; 17:2338-2347. [PMID: 34239360 PMCID: PMC8241723 DOI: 10.7150/ijbs.60332] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/29/2021] [Indexed: 01/25/2023] Open
Abstract
Rationale: Growth differentiation factor-8 (GDF-8), also known as myostatin, belongs to the transforming growth factor-beta (TGF-β) superfamily. GDF-8 is expressed in the ovary and regulates various ovarian functions. Ovarian hyperstimulation syndrome (OHSS) is one of the most serious disorders during in vitro fertilization treatment. Aromatase, encoded by the CYP19A1 gene, is the enzyme that catalyzes the final step in estradiol (E2) biosynthesis. It has been demonstrated that high serum E2 levels are associated with the development of OHSS. However, the effects of GDF-8 on aromatase expression and its roles in the pathogenesis of OHSS remain unclear. Methods: The effect of GDF-8 on aromatase expression and the underlying mechanisms were explored by a series of in vitro experiments in primary human granulosa-lutein (hGL) and KGN cells. Rat OHSS model and human follicular fluid samples were used to examine the roles of the GDF-8 system in the pathogenesis of OHSS. Results: We demonstrate that GDF-8 stimulates aromatase expression and E2 production in hGL and KGN cells. In addition, TGF-β type I receptor ALK5-mediated SMAD2/3 signaling is required for GDF-8-induced aromatase expression and E2 production. Using a rat OHSS model, we show that the aromatase and GDF-8 levels are upregulated in the ovaries of OHSS rats. Blocking the function of ALK5 by the administration of its inhibitor, SB431542, alleviates OHSS symptoms and the upregulation of aromatase. Clinical results reveal that the protein levels of GDF-8 are upregulated in the follicular fluid of OHSS patients. Moreover, the expression of GDF-8 is increased in hGL cells of OHSS patients. Conclusions: This study helps to elucidate the mechanisms mediating the expression of aromatase in human granulosa cells, which may lead to the development of alternative therapeutic approaches for OHSS.
Collapse
Affiliation(s)
- Lanlan Fang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Yan
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sijia Wang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, New Territories, Hong Kong, China
| | - Yanjie Guo
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiran Li
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, New Territories, Hong Kong, China
| | - Qiongqiong Jia
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Han
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Boqun Liu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jung-Chien Cheng
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying-Pu Sun
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
Salem AM. Variation of Leptin During Menstrual Cycle and Its Relation to the Hypothalamic-Pituitary-Gonadal (HPG) Axis: A Systematic Review. Int J Womens Health 2021; 13:445-458. [PMID: 34007218 PMCID: PMC8121381 DOI: 10.2147/ijwh.s309299] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/19/2021] [Indexed: 12/23/2022] Open
Abstract
Recently, adipose tissue has been identified as endocrine organ in addition to its action as energy store; it produces a large number of biologically active mediators known as adipocytokines. Significantly, adipocytokines were found to be involved in the physiology of many body functions, including reproduction. The role of body weight, body fat compositions, and nutrition has been largely investigated using animal models and human studies. Malnutrition and/or abnormal body weight may induce disturbances in fertility, puberty, pregnancy, and menstrual cycles. Leptin was the first discovered adipocytokine, and a large body of data over the last 25 years has shown that leptin is not only a molecule that reflects energy stores in the body, but is also an important cytokine involved in many physiological functions, such as inflammatory response, insulin sensitivity, bone metabolism, immunity, and most importantly, reproductive function. Leptin controls the normal physiology of the female reproductive system; it interacts with the hypothalamic–pituitary–gonadal (HPG) axis by a complex mechanism that connects energy homeostasis with reproduction. However, observational studies have demonstrated inconsistent results about leptin variation during normal menstrual cycle, and the mechanisms involved in the interplay between leptin and the hormones of the HPG axis are largely unknown. This review focuses on leptin variation during normal menstrual cycles and its relation to the hypothalamic–pituitary–gonadal axis, and the effect of overweight/obesity on leptin during menstrual cycle is further reviewed.
Collapse
Affiliation(s)
- Ayad Mohammed Salem
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
11
|
Weis-Banke SE, Lerdrup M, Kleine-Kohlbrecher D, Mohammad F, Sidoli S, Jensen ON, Yanase T, Nakamura T, Iwase A, Stylianou A, Abu-Rustum NR, Aghajanian C, Soslow R, Da Cruz Paula A, Koche RP, Weigelt B, Christensen J, Helin K, Cloos PAC. Mutant FOXL2 C134W Hijacks SMAD4 and SMAD2/3 to Drive Adult Granulosa Cell Tumors. Cancer Res 2020; 80:3466-3479. [PMID: 32641411 DOI: 10.1158/0008-5472.can-20-0259] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/26/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022]
Abstract
The mutant protein FOXL2C134W is expressed in at least 95% of adult-type ovarian granulosa cell tumors (AGCT) and is considered to be a driver of oncogenesis in this disease. However, the molecular mechanism by which FOXL2C134W contributes to tumorigenesis is not known. Here, we show that mutant FOXL2C134W acquires the ability to bind SMAD4, forming a FOXL2C134W/SMAD4/SMAD2/3 complex that binds a novel hybrid DNA motif AGHCAHAA, unique to the FOXL2C134W mutant. This binding induced an enhancer-like chromatin state, leading to transcription of nearby genes, many of which are characteristic of epithelial-to-mesenchymal transition. FOXL2C134W also bound hybrid loci in primary AGCT. Ablation of SMAD4 or SMAD2/3 resulted in strong reduction of FOXL2C134W binding at hybrid sites and decreased expression of associated genes. Accordingly, inhibition of TGFβ mitigated the transcriptional effect of FOXL2C134W. Our results provide mechanistic insight into AGCT pathogenesis, identifying FOXL2C134W and its interaction with SMAD4 as potential therapeutic targets to this condition. SIGNIFICANCE: FOXL2C134W hijacks SMAD4 and leads to the expression of genes involved in EMT, stemness, and oncogenesis in AGCT, making FOXL2C134W and the TGFβ pathway therapeutic targets in this condition. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/17/3466/F1.large.jpg.
Collapse
Affiliation(s)
- Stine E Weis-Banke
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen N, Denmark.,The Novo Nordisk Foundation Center for Stem Cell Research (DanStem), University of Copenhagen, Copenhagen N, Denmark
| | - Mads Lerdrup
- Center for Chromosome Stability, University of Copenhagen, Copenhagen N, Denmark
| | - Daniela Kleine-Kohlbrecher
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen N, Denmark.,The Novo Nordisk Foundation Center for Stem Cell Research (DanStem), University of Copenhagen, Copenhagen N, Denmark
| | - Faizaan Mohammad
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen N, Denmark.,The Novo Nordisk Foundation Center for Stem Cell Research (DanStem), University of Copenhagen, Copenhagen N, Denmark
| | - Simone Sidoli
- Department of Biochemistry and Molecular Biology, VILLUM Centre for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark.,Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Ole N Jensen
- Department of Biochemistry and Molecular Biology, VILLUM Centre for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Toshihiko Yanase
- Seiwakai Muta Hospital, 3-9-1 Hoshikuma, Sawara-ku, Fukuoka, Japan
| | - Tomoko Nakamura
- Departments of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Akira Iwase
- Department of Obstetrics and Gynecology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Anthe Stylianou
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nadeem R Abu-Rustum
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert Soslow
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Arnaud Da Cruz Paula
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jesper Christensen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen N, Denmark.,The Novo Nordisk Foundation Center for Stem Cell Research (DanStem), University of Copenhagen, Copenhagen N, Denmark
| | - Kristian Helin
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen N, Denmark. .,The Novo Nordisk Foundation Center for Stem Cell Research (DanStem), University of Copenhagen, Copenhagen N, Denmark.,Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul A C Cloos
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen N, Denmark. .,The Novo Nordisk Foundation Center for Stem Cell Research (DanStem), University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
12
|
Newly Identified Regulators of Ovarian Folliculogenesis and Ovulation. Int J Mol Sci 2020; 21:ijms21124565. [PMID: 32604954 PMCID: PMC7349727 DOI: 10.3390/ijms21124565] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
Each follicle represents the basic functional unit of the ovary. From its very initial stage of development, the follicle consists of an oocyte surrounded by somatic cells. The oocyte grows and matures to become fertilizable and the somatic cells proliferate and differentiate into the major suppliers of steroid sex hormones as well as generators of other local regulators. The process by which a follicle forms, proceeds through several growing stages, develops to eventually release the mature oocyte, and turns into a corpus luteum (CL) is known as “folliculogenesis”. The task of this review is to define the different stages of folliculogenesis culminating at ovulation and CL formation, and to summarize the most recent information regarding the newly identified factors that regulate the specific stages of this highly intricated process. This information comprises of either novel regulators involved in ovarian biology, such as Ube2i, Phoenixin/GPR73, C1QTNF, and α-SNAP, or recently identified members of signaling pathways previously reported in this context, namely PKB/Akt, HIPPO, and Notch.
Collapse
|
13
|
Zhang H, Qin F, Liu A, Sun Q, Wang Q, Li Q, Lu S, Zhang D, Lu Z. Kuntai capsule attenuates premature ovarian failure through the PI3K/AKT/mTOR pathway. JOURNAL OF ETHNOPHARMACOLOGY 2019; 239:111885. [PMID: 31009706 DOI: 10.1016/j.jep.2019.111885] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kuntai capsule (KTC), a type of herb formulas, was first described in the book of Shang Han Za Bing Lun in the third century. KTC has been widely used for the clinical treatment of menopausal syndrome. Considering that premature ovarian failure is also known as premature menopause, this study was designed to investigate the effects and mechanisms of KTC on a mouse model of premature ovarian failure. MATERIALS AND METHODS Forty-five female C57BL/6 mice were chosen for this study. Fifteen of the mice were separated into the Control group. The remaining thirty were used to establish the premature ovarian failure model by injecting intraperitoneally with 75 mg/kg cyclophosphamide and then by randomly dividing the mice into two groups. One group was considered the Model group, the other group treated with the Kuntai capsule intragastrically every day for one week called the KTC group. After treatment, mice were sacrificed for sampling. The ovaries morphology of mice was observed by hematoxylin and eosin (HE) staining, and all follicles were counted under microscope. Western blotting was used to detect the PI3K/AKT/mTOR pathway activation. Serum follicle-stimulating hormone (FSH), luteinizing hormone (LH), estradiol (E2) and anti-mullerian hormone (AMH)levels were measured by enzyme-linked immunosorbent assay (ELISA). The fertility was observed by giving treated mice 8 weeks for breeding. RESULTS We found that primordial follicle counts were increased in the KTC group compared to the Model group. The phosphorylation of PI3K, AKT, mTOR, 4E-BP1 and S6K in the KTC group significantly reduced compared to Model group. Serum FSH and LH levels in the KTC group were decreased compared to the Model group, while, serum E2 and AMH levels in the KTC group were increased compared with the Model group. The litter size in the KTC group was improved compared to Model group. CONCLUSIONS The KTC showed protective potentials of ovarian reserve and fertility to attenuate premature ovarian failure, which was relatively associated with activation of the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Han Zhang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Fenfen Qin
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China; College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Anlong Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qinmei Sun
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qisheng Wang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qian Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Second Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shengfeng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Second Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Dong Zhang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, 210093, China
| | - Zhigang Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China; College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
14
|
Haraguchi H, Hirota Y, Saito-Fujita T, Tanaka T, Shimizu-Hirota R, Harada M, Akaeda S, Hiraoka T, Matsuo M, Matsumoto L, Hirata T, Koga K, Wada-Hiraike O, Fujii T, Osuga Y. Mdm2-p53-SF1 pathway in ovarian granulosa cells directs ovulation and fertilization by conditioning oocyte quality. FASEB J 2019; 33:2610-2620. [PMID: 30260703 DOI: 10.1096/fj.201801401r] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Functions of tumor suppressor p53 and its negative regulator mouse double minute 2 homolog (Mdm2) in ovarian granulosa cells remain to be elucidated, and the current study aims at clarifying this issue. Mice with Mdm2 deficiency in ovarian granulosa cells [ Mdm2-loxP/ progesterone receptor ( Pgr)-Cre mice] were infertile as a result of impairment of oocyte maturation, ovulation, and fertilization, and those with Mdm2/p53 double deletion in granulosa cells ( Mdm2-loxP/ p53-loxP/ Pgr-Cre mice) showed normal fertility, suggesting that p53 induction in the ovarian granulosa cells is detrimental to ovarian function by disturbing oocyte quality. Another model of Mdm2 deletion in ovarian granulosa cells ( Mdm2-loxP/ anti-Mullerian hormone type 2 receptor-Cre mice) also showed subfertility as a result of the failure of ovulation and fertilization, indicating critical roles of ovarian Mdm2 in ovulation and fertilization. Mdm2-p53 pathway in cumulus granulosa cells transcriptionally controlled an orphan nuclear receptor steroidogenic factor 1 (SF1), a key regulator of ovarian function. Importantly, MDM2 and SF1 levels in human cumulus granulosa cells were positively associated with the outcome of oocyte maturation and fertilization in patients undergoing infertility treatment. These findings suggest that the Mdm2-p53-SF1 axis in ovarian cumulus granulosa cells directs ovarian function by affecting their neighboring oocyte quality.-Haraguchi, H., Hirota, Y., Saito-Fujita, T., Tanaka, T., Shimizu-Hirota, R., Harada, M., Akaeda, S., Hiraoka, T., Matsuo, M., Matsumoto, L., Hirata, T., Koga, K., Wada-Hiraike, O., Fujii, T., Osuga, Y. Mdm2-p53-SF1 pathway in ovarian granulosa cells directs ovulation and fertilization by conditioning oocyte quality.
Collapse
Affiliation(s)
- Hirofumi Haraguchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama, Japan
- Precursory Research for Innovative Medical Care, Japan Agency for Medical Research and Development, Tokyo, Japan; and
| | - Tomoko Saito-Fujita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoki Tanaka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryoko Shimizu-Hirota
- Department of Internal Medicine, Center for Preventive Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Miyuki Harada
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shun Akaeda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takehiro Hiraoka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsunori Matsuo
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Leona Matsumoto
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuya Hirata
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kaori Koga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Xia H, Zhang R, Guan H, Zhang W. Follicle loss and PTEN/PI3K/mTOR signaling pathway activated in LepR-mutated mice. Gynecol Endocrinol 2019; 35:44-48. [PMID: 30145913 DOI: 10.1080/09513590.2018.1490714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Female mice (Y123F) with substitution mutations introduced through homologous gene targeting, replacing the three tyrosine residues of LepR, Tyr985, Tyr1077, and Tyr1138 with phenylalanine, could induce infertility. This study aimed to describe the reproductive alteration and to explore its mechanism. We compared the reproductive characteristics in the female homozygous (HOM) Y123F mice and wild-type (WT) littermates, analyzing the expression of downstream molecules of LepR, like protein kinase B (Akt)/mammalian target of rapamycin (mTOR), phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and insulin receptor substrate (IRS) in the ovaries. The results showed that 10-week old female Y123F HOM exhibited no reproductive periods, declined anti-mullerian hormone (AMH) levels in the serum and ovaries, reduced primordial follicles, primary follicles, secondary follicles, antral follicles and hardly no corpus lutea (all p < .05). The phosphorylation of downsream Akt, mTOR, S6K1 and eIF4B of LepR were all elevated in the ovaries of the mutated female mice. They also presented a decreased phosphorylation of IRS-1, IRS-2, and PTEN, and a strengthened phosphorylation of FOXO-3A in the ovaries. In conclusions, LepR mutation could result in follicle loss and activation of PTEN/PI3K/Akt/mTOR pathway in adult female mice, independent of insulin signaling pathway.
Collapse
Affiliation(s)
- Hexia Xia
- a Department of Reproductive Endocrinology, Obstetrics and Gynecology Hospital , Fudan University , Shanghai , People's Republic of China
- b Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases , Shanghai , People's Republic of China
| | - Ruixiu Zhang
- a Department of Reproductive Endocrinology, Obstetrics and Gynecology Hospital , Fudan University , Shanghai , People's Republic of China
- b Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases , Shanghai , People's Republic of China
| | - Haiyun Guan
- a Department of Reproductive Endocrinology, Obstetrics and Gynecology Hospital , Fudan University , Shanghai , People's Republic of China
- b Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases , Shanghai , People's Republic of China
| | - Wei Zhang
- a Department of Reproductive Endocrinology, Obstetrics and Gynecology Hospital , Fudan University , Shanghai , People's Republic of China
- b Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases , Shanghai , People's Republic of China
| |
Collapse
|
16
|
Rafique N, Salem AM, Latif R, ALSheikh MH. Serum leptin level across different phases of menstrual cycle in normal weight and overweight/obese females. Gynecol Endocrinol 2018; 34:601-604. [PMID: 29268651 DOI: 10.1080/09513590.2017.1419173] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
We compared serum leptin levels during various phases of menstrual cycle and its correlation with serum estradiol between normal weight and overweight/obese young females. Fifty-six young females with normal menstrual cycle were grouped into 26 normal weight and 30 overweight/obese subjects. Serum leptin and estradiol levels were measured during early follicular, pre-ovulatory and luteal phases of menstrual cycle in both groups using ELISA technique. Serum leptin levels were significantly different across different phases of menstrual cycle with a steady increment from follicular phase (9.97 ± 5.48 ng/dl) through pre-ovulatory phase (11.58 ± 6.49 ng/dl) with their peaks in luteal phase (12.52 ± 6.39 ng/dl, p < .001). Same pattern of change during menstrual phases was observed when the normal weight and overweight/obese group were analyzed separately. Serum leptin levels were significantly higher in overweight/obese group compared to normal weight subjects. In any of the study groups, leptin levels were not found to be correlated with estradiol level during different phases of menstrual cycle.
Collapse
Affiliation(s)
- Nazish Rafique
- a Department of Physiology , College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Ayad M Salem
- a Department of Physiology , College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Rabia Latif
- a Department of Physiology , College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Mona H ALSheikh
- a Department of Physiology , College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| |
Collapse
|
17
|
Brown JL, Xie J, Brieño-Enriquez MA, Sones J, Angulo CN, Boehm U, Miller A, Toufaily C, Wang Y, Bernard DJ, Roberson MS. Sex- and Age-Specific Impact of ERK Loss Within the Pituitary Gonadotrope in Mice. Endocrinology 2018; 159:1264-1276. [PMID: 29300908 PMCID: PMC5802804 DOI: 10.1210/en.2017-00653] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/28/2017] [Indexed: 02/06/2023]
Abstract
Extracellular signal-regulated kinase (ERK) signaling regulates hormone action in the reproductive axis, but specific mechanisms have yet to be completely elucidated. In the current study, ERK1 null and ERK2 floxed mice were combined with a gonadotropin-releasing hormone receptor (GnRHR)-internal ribosomal entry site-Cre (GRIC) driver. Female ERK double-knockout (ERKdko) animals were hypogonadotropic, resulting in anovulation and complete infertility. Transcript levels of four gonadotrope-specific genes (GnRHR and the three gonadotropin subunits) were reduced in pituitaries at estrus in ERKdko females, and the postcastration response to endogenous GnRH hyperstimulation was blunted. As females aged, they exhibited abnormal ovarian histology, as well as increased body weight. ERKdko males were initially less affected, showing moderate subfertility, up to 6 months of age. Male ERKdko mice also displayed a blunted response to endogenous GnRH following castration. By 12 months of age, ERKdko males had reduced testicular weights and sperm production. By 18 months of age, the ERKdko males displayed reduced testis and seminal vesicle weights, marked seminiferous tubule degeneration, and a 77% reduction in sperm production relative to controls. As the GRIC is also active in the male germ line, we examined the specific role of ERK loss in the testes using the stimulated by retinoic acid 8 (Stra8)-Cre driver. Whereas ERK loss in GRIC and Stra8 males resulted in comparable losses in sperm production, seminiferous tubule histological degeneration was only observed in the GRIC-ERKdko animals. Our data suggest that loss of ERK signaling and hypogonadotropism within the reproductive axis impacts fertility and gonadal aging.
Collapse
Affiliation(s)
- Jessica L Brown
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Jianjun Xie
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | | | - Jennifer Sones
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, New York
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Cynthia N Angulo
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg, Germany
| | - Andrew Miller
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Chirine Toufaily
- Department of Pharmacology and Therapeutics, McGill University, Québec, Canada
| | - Ying Wang
- Department of Pharmacology and Therapeutics, McGill University, Québec, Canada
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Québec, Canada
| | - Mark S Roberson
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, New York
- Correspondence: Mark S. Roberson, PhD, Department of Biomedical Sciences, T4-018 Veterinary Research Tower, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853. E-mail:
| |
Collapse
|
18
|
Ulloa-Aguirre A, Zariñán T, Gutiérrez-Sagal R, Dias JA. Intracellular Trafficking of Gonadotropin Receptors in Health and Disease. Handb Exp Pharmacol 2018; 245:1-39. [PMID: 29063275 DOI: 10.1007/164_2017_49] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Gonadotropin receptors belong to the highly conserved subfamily of the G protein-coupled receptor (GPCR) superfamily, the so-called Rhodopsin-like family (class A), which is the largest class of GPCRs and currently a major drug target. Both the follicle-stimulating hormone receptor (FSHR) and the luteinizing hormone/chorionic gonadotropin hormone receptor (LHCGR) are mainly located in the gonads where they play key functions associated to essential reproductive functions. As any other protein, gonadotropin receptors must be properly folded into a mature tertiary conformation compatible with quaternary assembly and endoplasmic reticulum export to the cell surface plasma membrane. Several primary and secondary structural features, including presence of particular amino acid residues and short motifs and in addition, posttranslational modifications, regulate intracellular trafficking of gonadotropin receptors to the plasma membrane as well as internalization and recycling of the receptor back to the cell surface after activation by agonist. Inactivating mutations of gonadotropin receptors may derive from receptor misfolding and lead to absent or reduced plasma membrane expression of the altered receptor, thereby manifesting an array of phenotypical abnormalities mostly characterized by reproductive failure and/or abnormal or absence of development of secondary sex characteristics. In this chapter we review the structural requirements necessary for intracellular trafficking of the gonadotropin receptors, and describe how mutations in these receptors may lead to receptor misfolding and disease in humans.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición SZ, Vasco de Quiroga 15, Tlalpan, Mexico City, 14000, Mexico.
| | - Teresa Zariñán
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición SZ, Vasco de Quiroga 15, Tlalpan, Mexico City, 14000, Mexico
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición SZ, Vasco de Quiroga 15, Tlalpan, Mexico City, 14000, Mexico
| | - James A Dias
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, USA
| |
Collapse
|
19
|
Jayaraman A, Kumar TR. Extra-pituitary expressed follicle-stimulating hormone: Is it physiologically important? Biol Reprod 2017; 97:622-626. [PMID: 29036567 DOI: 10.1093/biolre/iox117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/19/2017] [Indexed: 11/14/2022] Open
Abstract
Pituitary gonadotropes synthesize and secrete follicle-stimulating hormone (FSH). FSH is a heterodimer that consists of an α- and β-subunit. The α-subunit is common to other pituitary and placental glycoprotein hormones, and the β-subunit is the hormone/receptor-specific subunit. Although the pituitary is the main tissue that accounts for circulating hormone, previous and recent reports indicate extra-pituitary sources of FSH production including mouse gonads, human stomach, prostate, umbilical cord vein endothelial cells, uterine myometrium, placenta, and chicken abdominal adipose tissue. Whether extra-pituitary derived FSH exerts any physiologically significant actions is not known. In this review, we have comprehensively analyzed the expression of mRNAs that encode mouse and human FSH subunits and also their corresponding expressed sequence tags in normal tissues, cancer cell lines, and primary tumors by public database mining. We propose criteria to assess the significance of individual FSH subunit or FSH dimer expression as well as genetic approaches to unambiguously define the physiological relevance of extra-pituitary FSH expression.
Collapse
Affiliation(s)
- Anushka Jayaraman
- Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - T Rajendra Kumar
- Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Program in Integrated Physiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Division of Reproductive Endocrinology & Infertility, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
20
|
Wang H, May J, Butnev V, Shuai B, May JV, Bousfield GR, Kumar TR. Evaluation of in vivo bioactivities of recombinant hypo- (FSH 21/18) and fully- (FSH 24) glycosylated human FSH glycoforms in Fshb null mice. Mol Cell Endocrinol 2016; 437:224-236. [PMID: 27561202 PMCID: PMC5048586 DOI: 10.1016/j.mce.2016.08.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/21/2016] [Accepted: 08/21/2016] [Indexed: 10/21/2022]
Abstract
The hormone - specific FSHβ subunit of the human FSH heterodimer consists of N-linked glycans at Asn7 and Asn24 residues that are co-translationally attached early during subunit biosynthesis. Differences in the number of N-glycans (none, one or two) on the human FSHβ subunit contribute to macroheterogeneity in the FSH heterodimer. The resulting FSH glycoforms are termed hypo-glycosylated (FSH21/18, missing either an Asn24 or Asn7 N-glycan chain on the β - subunit, respectively) or fully glycosylated (FSH24, possessing of both Asn7 and Asn24 N-linked glycans on the β - subunit) FSH. The recombinant versions of human FSH glycoforms (FSH21/18 and FSH24) have been purified and biochemically characterized. In vitro functional studies have indicated that FSH21/18 exhibits faster FSH- receptor binding kinetics and is much more active than FSH24 in every assay tested to date. However, the in vivo bioactivity of the hypo-glycosylated FSH glycoform has never been tested. Here, we evaluated the in vivo bioactivities of FSH glycoforms in Fshb null mice using a pharmacological rescue approach. In Fshb null female mice, both hypo- and fully-glycosylated FSH elicited an ovarian weight gain response by 48 h and induced ovarian genes in a dose- and time-dependent manner. Quantification by real time qPCR assays indicated that hypo-glycosylated FSH21/18 was bioactive in vivo and induced FSH-responsive ovarian genes similar to fully-glycosylated FSH24. Western blot analyses followed by densitometry of key signaling components downstream of the FSH-receptor confirmed that the hypo-glycosylated FSH21/18 elicited a response similar to that by fully-glycosylated FSH24 in ovaries of Fshb null mice. When injected into Fshb null males, hypo-glycosylated FSH21/18 was more active than the fully-glycosylated FSH24 in inducing FSH-responsive genes and Sertoli cell proliferation. Thus, our data establish that recombinant hypo-glycosylated human FSH21/18 glycoform elicits bioactivity in vivo similar to the fully-glycosylated FSH. Our studies may have clinical implications particularly in formulating FSH-based ovarian follicle induction protocols using a combination of different human FSH glycoforms.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jacob May
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Viktor Butnev
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260, USA
| | - Bin Shuai
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260, USA
| | - Jeffrey V May
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260, USA
| | - George R Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260, USA
| | - T Rajendra Kumar
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Center for Reproductive Sciences, Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Neurosurgery, University of Kansas Medical Center, Kansas City, KS 66160, USA; Division of Reproductive Sciences, Department of Obstetrics & Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
21
|
Mouse Models for the Study of Synthesis, Secretion, and Action of Pituitary Gonadotropins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:49-84. [PMID: 27697204 DOI: 10.1016/bs.pmbts.2016.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Gonadotropins play fundamental roles in reproduction. More than 30years ago, Cga transgenic mice were generated, and more than 20years ago, the phenotypes of Cga null mice were reported. Since then, numerous mouse strains have been generated and characterized to address several questions in reproductive biology involving gonadotropin synthesis, secretion, and action. More recently, extragonadal expression, and in some cases, functions of gonadotropins in nongonadal tissues have been identified. Several genomic and proteomic approaches including novel mouse genome editing tools are available now. It is anticipated that these and other emerging technologies will be useful to build an integrated network of gonadotropin signaling pathways in various tissues. Undoubtedly, research on gonadotropins will continue to provide new knowledge and allow us transcend from benchside to the bedside.
Collapse
|
22
|
Toda K, Hayashi Y, Ono M, Saibara T. Characterization of Ovarian Responses to Equine Chorionic Gonadotropin of Aromatase-Deficient Mice With or Without 17β-Estradiol Supplementation. Endocrinology 2016; 157:2093-103. [PMID: 26919384 DOI: 10.1210/en.2015-1701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aromatase is an enzyme catalyzing the final step of 17β-estradiol (E2) biosynthesis. Aromatase-deficient (ArKO) mice displayed vital roles of E2 at various tissue sites, including ovary. Here, we report attenuated responses of ArKO ovary to equine chorionic gonadotropin (eCG), an alternative to FSH. Ovarian contents of cAMP and anti-Müllerian hormone (AMH), putative factors reducing sensitivity to gonadotropins, were significantly elevated in ArKO mice compared with those in wild type (WT) mice in the basal state. Accordingly, eCG-induced ovarian alterations in cAMP contents, phosphorylation levels of signaling molecules, and mRNA expression of eCG-targeted genes were blunted in ArKO mice compared with those in WT mice. Treatment of ArKO mice with E2 decreased ovarian cAMP and AMH contents to the WT levels but did not restore the sensitivity. Microarray analysis coupled with quantitative RT-PCR analysis identified 7 genes of which the mRNA expression levels in ArKO ovaries were significantly different from those in the WT ovaries in the basal state and were not normalized by E2 supplementation, indicating possible involvement of these gene products in the determination of ovarian sensitivity to eCG. Thus, present analyses revealed that estrogen deficiency attenuates sensitivity of the ovary to gonadotropin, which might be associated with alterations in the ovarian contents of multiple molecules including cAMP and AMH. Given the importance of the ovarian responses to gonadotropins in reproductive function, detailed knowledge about the underlying mechanisms of abnormalities in the ArKO ovary might help to develop potential targets for infertility treatments.
Collapse
Affiliation(s)
- Katsumi Toda
- Departments of Biochemistry (K.T.), Pathology (Y.H.), and Gastroenterology and Hepatology (M.O., T.S.), Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Yoshihiro Hayashi
- Departments of Biochemistry (K.T.), Pathology (Y.H.), and Gastroenterology and Hepatology (M.O., T.S.), Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Masafumi Ono
- Departments of Biochemistry (K.T.), Pathology (Y.H.), and Gastroenterology and Hepatology (M.O., T.S.), Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Toshiji Saibara
- Departments of Biochemistry (K.T.), Pathology (Y.H.), and Gastroenterology and Hepatology (M.O., T.S.), Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| |
Collapse
|
23
|
Binder AK, Kosak JP, Janhardhan KS, Moser G, Eling TE, Korach KS. Expression of Human NSAID Activated Gene 1 in Mice Leads to Altered Mammary Gland Differentiation and Impaired Lactation. PLoS One 2016; 11:e0146518. [PMID: 26745373 PMCID: PMC4706436 DOI: 10.1371/journal.pone.0146518] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/19/2015] [Indexed: 12/22/2022] Open
Abstract
Transgenic mice expressing human non-steroidal anti-inflammatory drug activated gene 1 (NAG-1) have less adipose tissue, improved insulin sensitivity, lower insulin levels and are resistant to dietary induced obesity. The hNAG-1 expressing mice are more metabolically active with a higher energy expenditure. This study investigates female reproduction in the hNAG-1 transgenic mice and finds the female mice are fertile but have reduced pup survival after birth. Examination of the mammary glands in these mice suggests that hNAG-1 expressing mice have altered mammary epithelial development during pregnancy, including reduced occupancy of the fat pad and increased apoptosis via TUNEL positive cells on lactation day 2. Pups nursing from hNAG-1 expressing dams have reduced milk spots compared to pups nursing from WT dams. When CD-1 pups were cross-fostered with hNAG-1 or WT dams; reduced milk volume was observed in pups nursing from hNAG-1 dams compared to pups nursing from WT dams in a lactation challenge study. Milk was isolated from WT and hNAG-1 dams, and the milk was found to have secreted NAG-1 protein (approximately 25 ng/mL) from hNAG-1 dams. The WT dams had no detectable hNAG-1 in the milk. A decrease in non-esterified free fatty acids in the milk of hNAG-1 dams was observed. Altered milk composition suggests that the pups were receiving inadequate nutrients during perinatal development. To examine this hypothesis serum was isolated from pups and clinical chemistry points were measured. Male and female pups nursing from hNAG-1 dams had reduced serum triglyceride concentrations. Microarray analysis revealed that genes involved in lipid metabolism are differentially expressed in hNAG-1 mammary glands. Furthermore, the expression of Cidea/CIDEA that has been shown to regulate milk lipid secretion in the mammary gland was reduced in hNAG-1 mammary glands. This study suggests that expression of hNAG-1 in mice leads to impaired lactation and reduces pup survival due to altered milk quality and quantity.
Collapse
Affiliation(s)
- April K. Binder
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Justin P. Kosak
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Kyathanahalli S. Janhardhan
- Integrated Laboratory Systems Incorporated, Research Triangle Park, North Carolina, United States of America
| | - Glenda Moser
- Integrated Laboratory Systems Incorporated, Research Triangle Park, North Carolina, United States of America
| | - Thomas E. Eling
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Kenneth S. Korach
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
24
|
del Pino J, Moyano-Cires PV, Anadon MJ, Díaz MJ, Lobo M, Capo MA, Frejo MT. Molecular Mechanisms of Amitraz Mammalian Toxicity: A Comprehensive Review of Existing Data. Chem Res Toxicol 2015; 28:1073-94. [PMID: 25973576 DOI: 10.1021/tx500534x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Javier del Pino
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Paula Viviana Moyano-Cires
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Maria Jose Anadon
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - María Jesús Díaz
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Margarita Lobo
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Miguel Andrés Capo
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - María Teresa Frejo
- Departament of Toxicology
and Pharmacology, Veterinary School, and ‡Department of
Toxicology and Legal Medicine, Medicine School, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| |
Collapse
|
25
|
Chen Z, Kang X, Wang L, Dong H, Wang C, Xiong Z, Zhao W, Jia C, Lin J, Zhang W, Yuan W, Zhong M, Du H, Bai X. Rictor/mTORC2 pathway in oocytes regulates folliculogenesis, and its inactivation causes premature ovarian failure. J Biol Chem 2015; 290:6387-96. [PMID: 25564616 DOI: 10.1074/jbc.m114.605261] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Molecular basis of ovarian folliculogenesis and etiopathogenesis of premature ovarian failure (POF), a common cause of infertility in women, are not fully understood. Mechanistic target of rapamycin complex 2 (mTORC2) is emerging as a central regulator of cell metabolism, proliferation, and survival. However, its role in folliculogenesis and POF has not been reported. Here, we showed that the signaling activity of mTORC2 is inhibited in a 4-vinylcyclohexene diepoxide (VCD)-induced POF mouse model. Notably, mice with oocyte-specific ablation of Rictor, a key component of mTORC2, demonstrated POF phenotypes, including massive follicular death, excessive loss of functional ovarian follicles, abnormal gonadal hormone secretion, and consequently, secondary subfertility in conditional knock-out (cKO) mice. Furthermore, reduced levels of Ser-473-phosphorylated Akt and Ser-253-phosphorylated Foxo3a and elevated pro-apoptotic proteins, Bad, Bax, and cleaved poly ADP-ribose polymerase (PARP), were observed in cKO mice, replicating the signaling alterations in 4-VCD-treated ovaries. These results indicate a critical role of the Rictor/mTORC2/Akt/Foxo3a pro-survival signaling axis in folliculogenesis. Interestingly, loss of maternal Rictor did not cause obvious developmental defects in embryos or placentas from cKO mice, suggesting that maternal Rictor is dispensable for preimplantation embryonic development. Our results collectively indicate key roles of Rictor/mTORC2 in folliculogenesis, follicle survival, and female fertility and support the utility of oocyte-specific Rictor knock-out mice as a novel model for POF.
Collapse
Affiliation(s)
- Zhenguo Chen
- From the State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515
| | - Xiangjin Kang
- the Center for Reproductive Medicine, Third Affiliated Hospital of Guangzhou Medical University, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, and Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou 510150, and
| | - Liping Wang
- the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, and
| | - Heling Dong
- From the State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515
| | - Caixia Wang
- From the State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515
| | - Zhi Xiong
- From the State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515
| | - Wanlu Zhao
- From the State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515
| | - Chunhong Jia
- From the State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515
| | - Jun Lin
- From the State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515
| | - Wen Zhang
- the Center for Reproductive Medicine, Third Affiliated Hospital of Guangzhou Medical University, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, and Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou 510150, and
| | - Weiping Yuan
- the State Key Laboratory of Experimental Hematology, Institute of Hematology, Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Mei Zhong
- the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, and
| | - Hongzi Du
- the Center for Reproductive Medicine, Third Affiliated Hospital of Guangzhou Medical University, Key Laboratory for Reproductive Medicine of Guangdong Province, Key Laboratory for Major Obstetric Diseases of Guangdong Province, and Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou 510150, and
| | - Xiaochun Bai
- From the State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515,
| |
Collapse
|
26
|
Field SL, Dasgupta T, Cummings M, Orsi NM. Cytokines in ovarian folliculogenesis, oocyte maturation and luteinisation. Mol Reprod Dev 2013; 81:284-314. [DOI: 10.1002/mrd.22285] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/18/2013] [Indexed: 01/22/2023]
Affiliation(s)
- Sarah L Field
- Women's Health Research Group; Leeds Institute of Cancer; Anatomy and Pathology; Wellcome Trust Brenner Building; St James's University Hospital; Leeds UK
| | - Tathagata Dasgupta
- Department of Systems Biology; Harvard Medical School; 200 Longwood Avenue Boston Massachusetts
| | - Michele Cummings
- Women's Health Research Group; Leeds Institute of Cancer; Anatomy and Pathology; Wellcome Trust Brenner Building; St James's University Hospital; Leeds UK
| | - Nicolas M. Orsi
- Women's Health Research Group; Leeds Institute of Cancer; Anatomy and Pathology; Wellcome Trust Brenner Building; St James's University Hospital; Leeds UK
| |
Collapse
|
27
|
Ramalho-Santos J, Amaral S. Mitochondria and mammalian reproduction. Mol Cell Endocrinol 2013; 379:74-84. [PMID: 23769709 DOI: 10.1016/j.mce.2013.06.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/22/2013] [Accepted: 06/06/2013] [Indexed: 12/22/2022]
Abstract
Mitochondria are cellular organelles with crucial roles in ATP synthesis, metabolic integration, reactive oxygen species (ROS) synthesis and management, the regulation of apoptosis (namely via the intrinsic pathway), among many others. Additionally, mitochondria in different organs or cell types may have distinct properties that can decisively influence functional analysis. In terms of the importance of mitochondria in mammalian reproduction, and although there are species-specific differences, these aspects involve both energetic considerations for gametogenesis and fertilization, control of apoptosis to ensure the proper production of viable gametes, and ROS signaling, as well as other emerging aspects. Crucially, mitochondria are the starting point for steroid hormone biosynthesis, given that the conversion of cholesterol to pregnenolone (a common precursor for all steroid hormones) takes place via the activity of the cytochrome P450 side-chain cleavage enzyme (P450scc) on the inner mitochondrial membrane. Furthermore, mitochondrial activity in reproduction has to be considered in accordance with the very distinct strategies for gamete production in the male and female. These include distinct gonad morpho-physiologies, different types of steroids that are more prevalent (testosterone, estrogens, progesterone), and, importantly, the very particular timings of gametogenesis. While spermatogenesis is complete and continuous since puberty, producing a seemingly inexhaustible pool of gametes in a fixed environment; oogenesis involves the episodic production of very few gametes in an environment that changes cyclically. These aspects have always to be taken into account when considering the roles of any common element in mammalian reproduction.
Collapse
Affiliation(s)
- João Ramalho-Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Portugal.
| | | |
Collapse
|
28
|
Faustino L, Lima I, Carvalho A, Silva C, Castro S, Lobo C, Lucci C, Campello C, Figueiredo J, Rodrigues A. Interaction between keratinocyte growth factor-1 and kit ligand on the goat preantral follicles cultured in vitro. Small Rumin Res 2013. [DOI: 10.1016/j.smallrumres.2013.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
29
|
Toda K, Ono M, Yuhki KI, Ushikubi F, Saibara T. 17β-Estradiol is critical for the preovulatory induction of prostaglandin E(2) synthesis in mice. Mol Cell Endocrinol 2012; 362:176-82. [PMID: 22713853 DOI: 10.1016/j.mce.2012.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 06/08/2012] [Accepted: 06/12/2012] [Indexed: 10/28/2022]
Abstract
Aromatase-deficient (ArKO) mice are totally anovulatory due to insufficient estrogen production. However, sequential administrations of high doses of 17β-estradiol (E2) and gonadotropins were found to induce ovulation in these mice. Here, we examined how the ovulatory stimulation for ArKO mice alters the expressions of genes related to prostaglandin (PG) E(2) metabolism and ovarian contents of PGE(2), as PGE(2) is one of the critical mediators of ovulatory induction. The ovulatory stimulation significantly increased mRNA expressions of prostaglandin-endoperoxide synthase 2, PGE(2) receptor type 4 and sulfotransferase family 1E, member 1, in preovulatory ArKO ovaries. In contrast, it suppressed the mRNA expression of 15-hydroxyprostaglandin dehydrogenase. Furthermore, significant elevation in the PGE(2) contents was detected in the preovulatory ovaries of ArKO mice after stimulation with E2 plus ovulatory doses of gonadotropins. Thus, these analyses demonstrate a requirement of E2 for the preovulatory enhancement of PGE(2) synthesis, leading to future success in ovulation.
Collapse
Affiliation(s)
- Katsumi Toda
- Department of Biochemistry, Kochi University, School of Medicine, Nankoku, Kochi 783-8505, Japan.
| | | | | | | | | |
Collapse
|
30
|
Li F, Jo M, Curry TE, Liu J. Hormonal induction of polo-like kinases (Plks) and impact of Plk2 on cell cycle progression in the rat ovary. PLoS One 2012; 7:e41844. [PMID: 22870256 PMCID: PMC3411565 DOI: 10.1371/journal.pone.0041844] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 06/29/2012] [Indexed: 11/19/2022] Open
Abstract
The highly conserved polo-like kinases (Plks) are potent regulators of multiple functions in the cell cycle before and during mitotic cell division. We investigated the expression pattern of Plk genes and their potential role(s) in the rat ovary during the periovulatory period. Plk2 and Plk3 were highly induced both in intact ovaries and granulosa cells in vivo after treatment with the luteinizing hormone (LH) agonist, human chorionic gonadotropin (hCG). In vitro, hCG stimulated the expression of Plk2 in granulosa cells, but not Plk3. This induction of Plk2 expression was mimicked by both forskolin and phorbol 12 myristate 13-acetate (PMA). Moreover, Plk2 expression was reduced by inhibitors of prostaglandin synthesis or the EGF pathway, but not by progesterone receptor antagonist (RU486) treatment. At the promoter level, mutation of the Sp1 binding sequence abolished the transcriptional activity of the Plk2 gene. ChIP assays also revealed the interaction of endogenous Sp1 protein in the Plk2 promoter region. Functionally, the over-expression of Plk2 and Plk3 arrested granulosa cells at the G0/G1 phase of the cell cycle. In contrast, the knockdown of Plk2 expression in granulosa cells decreased the number of cells in the G0/G1 stage of the cell cycle, but increased granulosa cell viability. In summary, hCG induced Plk2 and Plk3 expression in the rat ovary. Prostaglandins and the EGF signaling pathway are involved in regulating Plk2 expression. The transcription factor Sp1 is important for Plk2 transcriptional up-regulation. Our findings suggest that the increase in Plk2 and Plk3 expression contributes to the cell cycle arrest of granulosa cells which is important for the luteinization of granulosa cells during the periovulatory period.
Collapse
Affiliation(s)
- Feixue Li
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, People’s Republic of China
| | - Misung Jo
- Department of Obstetrics and Gynecology, Chandler Medical Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Thomas E. Curry
- Department of Obstetrics and Gynecology, Chandler Medical Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jing Liu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, People’s Republic of China
- * E-mail:
| |
Collapse
|
31
|
Toda K, Hayashi Y, Ono M, Saibara T. Impact of ovarian sex steroids on ovulation and ovulatory gene induction in aromatase-null mice. Endocrinology 2012; 153:386-94. [PMID: 22147013 DOI: 10.1210/en.2011-1462] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Female mice deficient in the aromatase gene [aromatase knockout (ArKO)] fail to ovulate owing to an inability to produce estrogens. Here, we demonstrated that sequential administration of adequate amounts of 17β-estradiol (E2), pregnant mare serum gonadotropin, and human chorionic gonadotropin could induce ovulation in immature ArKO mice; nevertheless, significantly fewer oocytes were released into the oviducts in ArKO mice than in wild-type mice. Analysis of ovarian steroids by liquid chromatography coupled with electrospray ionization-tandem mass spectrometry identified a trace amount of E2 in the untreated immature ArKO ovary. The analysis further detected significant increases and decreases in progesterone and testosterone contents, respectively, in addition to an increase of E2 in the ovulation-induced ArKO ovaries compared with the levels in untreated ArKO ovaries. Gene expression analysis demonstrated marked elevation in the mRNA levels of members of the epidermal growth factor family and extracellular matrix-related proteins at 4 h after human chorionic gonadotropin injection in the ovaries of ArKO mice treated for ovulation, as observed in the ovulation-induced wild-type ovaries. Collectively, these findings suggest the vital contribution of the intraovarian milieu of sex steroids to ovulatory regulation in vivo.
Collapse
Affiliation(s)
- Katsumi Toda
- Department of Biochemistry, Kochi University, School of Medicine, Nankoku, Kochi 783-8505, Japan.
| | | | | | | |
Collapse
|
32
|
Conti M. Phosphodiesterases and regulation of female reproductive function. Curr Opin Pharmacol 2011; 11:665-9. [PMID: 22019564 DOI: 10.1016/j.coph.2011.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 09/29/2011] [Accepted: 10/04/2011] [Indexed: 01/19/2023]
Abstract
The function of the mammalian follicle is regulated by circulating gonadotropins and a myriad of local paracrine regulations functioning within the ovary. Cyclic nucleotide signaling plays an essential role in mediating both these endocrine and paracrine regulations. Given their role in controlling cyclic nucleotide levels, cyclic nucleotide phosphodiesterases (PDEs) are critical for ovarian function. PDEs modulate gonadotropin responses in the somatic compartment of the ovarian follicle. Specific PDEs are also essential for the oocyte entry and exit from the meiotic cell cycle. This review summarizes the function and regulation of PDEs in the somatic compartment and in the oocyte, and highlights the potential of PDEs as targets to improve assisted reproduction, as well as for fertility control.
Collapse
Affiliation(s)
- Marco Conti
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, 513 Parnassus Avenue, HSW 1656, Box 0556, San Francisco, CA 94143-0556, United States.
| |
Collapse
|