1
|
Feng F, Liu T, Hou X, Lin X, Zhou S, Tian Y, Qi X. Targeting the FSH/FSHR axis in ovarian cancer: advanced treatment using nanotechnology and immunotherapy. Front Endocrinol (Lausanne) 2024; 15:1489767. [PMID: 39741875 PMCID: PMC11685086 DOI: 10.3389/fendo.2024.1489767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Ovarian cancer (OC) is the gynecological malignancy with the poorest prognosis. Surgery and chemotherapy are the primary therapies for OC; however, patients often experience recurrence. Given the intimate interaction between OC cells and the tumor microenvironment (TME), it is imperative to devise treatments that target both tumor cells and TME components. Recently, follicle-stimulating hormone (FSH) levels in the blood have been shown to correlate with poorer prognosis in individuals with OC. Ovarian carcinoma cells express FSH receptors (FSHRs). Thus, FSH is an important target in the development of novel therapeutic agents. Here, we review the effects of FSH on normal physiology, including the reproductive, skeletal, cardiac, and fat metabolic systems. Importantly, this review outlines the role and mechanism of the FSH/FSHR axis in the proliferation, survival, and metastasis of OC, providing theoretical support for the targeted FSHR treatment of OC. Current progress in targeting FSHR for OC, including the recent application of nanotechnology and immunotherapy, is presented. Finally, we discuss prospects and future directions of targeted FSHR therapy in OC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaoyi Qi
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
2
|
Chacón C, Mounieres C, Ampuero S, Urzúa U. Transcriptomic Analysis of the Aged Nulliparous Mouse Ovary Suggests a Stress State That Promotes Pro-Inflammatory Lipid Signaling and Epithelial Cell Enrichment. Int J Mol Sci 2023; 25:513. [PMID: 38203684 PMCID: PMC10779227 DOI: 10.3390/ijms25010513] [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: 11/02/2023] [Revised: 12/23/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Ovarian cancer (OC) incidence and mortality peaks at post-menopause while OC risk is either reduced by parity or increased by nulliparity during fertile life. The long-term effect of nulliparity on ovarian gene expression is largely unknown. In this study, we describe a bioinformatic/data-mining analysis of 112 coding genes upregulated in the aged nulliparous (NP) mouse ovary compared to the aged multiparous one as reference. Canonical gene ontology and pathway analyses indicated a pro-oxidant, xenobiotic-like state accompanied by increased metabolism of inflammatory lipid mediators. Up-regulation of typical epithelial cell markers in the aged NP ovary was consistent with synchronized overexpression of Cldn3, Ezr, Krt7, Krt8 and Krt18 during the pre-neoplastic phase of mOSE cell cultures in a former transcriptome study. In addition, 61/112 genes were upregulated in knockout mice for Fshr and for three other tumor suppressor genes (Pten, Cdh1 and Smad3) known to regulate follicular homeostasis in the mammalian ovary. We conclude that the aged NP ovary displays a multifaceted stress state resulting from oxidative imbalance and pro-inflammatory lipid signaling. The enriched epithelial cell content might be linked to follicle depletion and is consistent with abundant clefts and cysts observed in aged human and mouse ovaries. It also suggests a mesenchymal-to-epithelial transition in the mOSE of the aged NP ovary. Our analysis suggests that in the long term, nulliparity worsens a variety of deleterious effects of aging and senescence thereby increasing susceptibility to cancer initiation in the ovary.
Collapse
Affiliation(s)
- Carlos Chacón
- Laboratorio de Genómica Aplicada, Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (C.C.); (C.M.)
| | - Constanza Mounieres
- Laboratorio de Genómica Aplicada, Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (C.C.); (C.M.)
| | - Sandra Ampuero
- Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Ulises Urzúa
- Laboratorio de Genómica Aplicada, Departamento de Oncología Básico Clínica, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (C.C.); (C.M.)
| |
Collapse
|
3
|
Caroppo E, Niederberger CS. Follicle-stimulating hormone treatment for male factor infertility. Fertil Steril 2023; 119:173-179. [PMID: 36470702 DOI: 10.1016/j.fertnstert.2022.09.362] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 12/11/2022]
Abstract
Follicle-stimulating hormone (FSH) treatment has been proven effective in stimulating spermatogenesis and improving the reproductive ability of men with hypogonadotropic hypogonadism, while the usefulness of such a treatment in infertile patients with normal pituitary function is restricted to a subgroup of responders that, however, cannot be identified by the current diagnostic tools before treatment. In this review we summarize the role played by FSH in the modulation of spermatogenesis, the effect of FSH treatment at a standard replacement dose and at higher dose on sperm parameters, spontaneous and in vitro fertilization pregnancy rates, and the efforts made to identify possible responders to FSH treatment.
Collapse
Affiliation(s)
- Ettore Caroppo
- Asl Bari, Reproductive Unit, Andrology Outpatients Clinics, Conversano, Italy.
| | - Craig S Niederberger
- Department of Urology, University of Illinois at Chicago, Chicago, Illinois; Department of Bioengineering, University of Illinois at Chicago College of Engineering, Chicago, Illinois
| |
Collapse
|
4
|
Male infertility and gonadotropin treatment: What can we learn from real-world data? Best Pract Res Clin Obstet Gynaecol 2023; 86:102310. [PMID: 36682942 DOI: 10.1016/j.bpobgyn.2022.102310] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 12/31/2022]
Abstract
Gonadotropin therapy to treat specific male infertility disorders associated with hypogonadotropic hypogonadism is evidence-based and effective in restoring spermatogenesis and fertility. In contrast, its use to improve fertility in men with idiopathic oligozoospermia or nonobstructive azoospermia remains controversial, despite being widely practiced. The existence of two major inter-related pathways for spermatogenesis, including FSH and intratesticular testosterone, provides a rationale for empiric hormone stimulation therapy in both eugonadal and hypogonadal males with idiopathic oligozoospermia or nonobstructive azoospermia. Real-world data (RWD) on gonadotropin stimulating for these patient subsets, mainly using human chorionic gonadotropin and follicle-stimulating hormone, accumulated gradually, showing a positive therapeutic effect in some patients, translated by increased sperm production, sperm quality, and sperm retrieval rates. Although more evidence is needed, current insights from RWD research indicate that selected male infertility patients might be managed more effectively using gonadotropin therapy, with potential gains for all parties involved.
Collapse
|
5
|
Wang JM, Li ZF, Yang WX, Tan FQ. Follicle-stimulating hormone signaling in Sertoli cells: a licence to the early stages of spermatogenesis. Reprod Biol Endocrinol 2022; 20:97. [PMID: 35780146 PMCID: PMC9250200 DOI: 10.1186/s12958-022-00971-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/20/2022] [Indexed: 11/10/2022] Open
Abstract
Follicle-stimulating hormone signaling is essential for the initiation and early stages of spermatogenesis. Follicle-stimulating hormone receptor is exclusively expressed in Sertoli cells. As the only type of somatic cell in the seminiferous tubule, Sertoli cells regulate spermatogenesis not only by controlling their own number and function but also through paracrine actions to nourish germ cells surrounded by Sertoli cells. After follicle-stimulating hormone binds to its receptor and activates the follicle-stimulating hormone signaling pathway, follicle-stimulating hormone signaling will establish a normal Sertoli cell number and promote their differentiation. Spermatogonia pool maintenance, spermatogonia differentiation and their entry into meiosis are also positively regulated by follicle-stimulating hormone signaling. In addition, follicle-stimulating hormone signaling regulates germ cell survival and limits their apoptosis. Our review summarizes the aforementioned functions of follicle-stimulating hormone signaling in Sertoli cells. We also describe the clinical potential of follicle-stimulating hormone treatment in male patients with infertility. Furthermore, our review may be helpful for developing better therapies for treating patients with dysfunctional follicle-stimulating hormone signaling in Sertoli cells.
Collapse
Affiliation(s)
- Jia-Ming Wang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhen-Fang Li
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Fu-Qing Tan
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
6
|
Haldar S, Agrawal H, Saha S, Straughn AR, Roy P, Kakar SS. Overview of follicle stimulating hormone and its receptors in reproduction and in stem cells and cancer stem cells. Int J Biol Sci 2022; 18:675-692. [PMID: 35002517 PMCID: PMC8741861 DOI: 10.7150/ijbs.63721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/21/2021] [Indexed: 11/05/2022] Open
Abstract
Follicle stimulating hormone (FSH) and its receptor (FSHR) have been reported to be responsible for several physiological functions and cancers. The responsiveness of stem cells and cancer stem cells towards the FSH-FSHR system make the function of FSH and its receptors more interesting in the context of cancer biology. This review is comprised of comprehensive information on FSH-FSHR signaling in normal physiology, gonadal stem cells, cancer cells, and potential options of utilizing FSH-FSHR system as an anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Swati Haldar
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand 247667, India.,Current address: Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand 249405
| | - Himanshu Agrawal
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand 247667, India
| | - Sarama Saha
- Department of Biochemistry, All India Institute of Medical Sciences Rishikesh, Uttarakhand 249203, India
| | - Alex R Straughn
- Department of Physiology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand 247667, India
| | - Sham S Kakar
- Department of Physiology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
7
|
Clément F, Crépieux P, Yvinec R, Monniaux D. Mathematical modeling approaches of cellular endocrinology within the hypothalamo-pituitary-gonadal axis. Mol Cell Endocrinol 2020; 518:110877. [PMID: 32569857 DOI: 10.1016/j.mce.2020.110877] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 01/26/2023]
Abstract
The reproductive neuroendocrine axis, or hypothalamo-pituitary-gonadal (HPG) axis, is a paragon of complex biological system involving numerous cell types, spread over several anatomical levels communicating through entangled endocrine feedback loops. The HPG axis exhibits remarkable dynamic behaviors on multiple time and space scales, which are an inexhaustible source of studies for mathematical and computational biology. In this review, we will describe a variety of modeling approaches of the HPG axis from a cellular endocrinology viewpoint. We will in particular investigate the questions raised by some of the most striking features of the HPG axis: (i) the pulsatile secretion of hypothalamic and pituitary hormones, and its counterpart, the cell signaling induced by frequency-encoded hormonal signals, and (ii) the dual, gametogenic and glandular function of the gonads, which relies on the tight control of the somatic cell populations ensuring the proper maturation and timely release of the germ cells.
Collapse
Affiliation(s)
- Frédérique Clément
- Inria, Centre de Recherche Inria Saclay-Île-de-France, Palaiseau, France.
| | - Pascale Crépieux
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France; CNRS, UMR7247, F-37380, Nouzilly, France; Université de Tours, F-37041, Tours, France
| | - Romain Yvinec
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France; CNRS, UMR7247, F-37380, Nouzilly, France; Université de Tours, F-37041, Tours, France
| | - Danielle Monniaux
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France; CNRS, UMR7247, F-37380, Nouzilly, France; Université de Tours, F-37041, Tours, France
| |
Collapse
|
8
|
Follicle-stimulating Hormone (FSH) Action on Spermatogenesis: A Focus on Physiological and Therapeutic Roles. J Clin Med 2020; 9:jcm9041014. [PMID: 32260182 PMCID: PMC7230878 DOI: 10.3390/jcm9041014] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human reproduction is regulated by the combined action of the follicle-stimulating hormone (FSH) and the luteinizing hormone (LH) on the gonads. Although FSH is largely used in female reproduction, in particular in women attending assisted reproductive techniques to stimulate multi-follicular growth, its efficacy in men with idiopathic infertility is not clearly demonstrated. Indeed, whether FSH administration improves fertility in patients with hypogonadotropic hypogonadism, the therapeutic benefit in men presenting alterations in sperm production despite normal FSH serum levels is still unclear. In the present review, we evaluate the potential pharmacological benefits of FSH administration in clinical practice. METHODS This is a narrative review, describing the FSH physiological role in spermatogenesis and its potential therapeutic action in men. RESULTS The FSH role on male fertility is reviewed starting from the physiological control of spermatogenesis, throughout its mechanism of action in Sertoli cells, the genetic regulation of its action on spermatogenesis, until the therapeutic options available to improve sperm production. CONCLUSION FSH administration in infertile men has potential benefits, although its action should be considered by evaluating its synergic action with testosterone, and well-controlled, powerful trials are required. Prospective studies and new compounds could be developed in the near future.
Collapse
|
9
|
Casarini L, Crépieux P, Reiter E, Lazzaretti C, Paradiso E, Rochira V, Brigante G, Santi D, Simoni M. FSH for the Treatment of Male Infertility. Int J Mol Sci 2020; 21:ijms21072270. [PMID: 32218314 PMCID: PMC7177393 DOI: 10.3390/ijms21072270] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022] Open
Abstract
Follicle-stimulating hormone (FSH) supports spermatogenesis acting via its receptor (FSHR), which activates trophic effects in gonadal Sertoli cells. These pathways are targeted by hormonal drugs used for clinical treatment of infertile men, mainly belonging to sub-groups defined as hypogonadotropic hypogonadism or idiopathic infertility. While, in the first case, fertility may be efficiently restored by specific treatments, such as pulsatile gonadotropin releasing hormone (GnRH) or choriogonadotropin (hCG) alone or in combination with FSH, less is known about the efficacy of FSH in supporting the treatment of male idiopathic infertility. This review focuses on the role of FSH in the clinical approach to male reproduction, addressing the state-of-the-art from the little data available and discussing the pharmacological evidence. New compounds, such as allosteric ligands, dually active, chimeric gonadotropins and immunoglobulins, may represent interesting avenues for future personalized, pharmacological approaches to male infertility.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-0593961705; Fax: +39-0593962018
| | - Pascale Crépieux
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Français du Cheval et de l’Equitation (IFCE), Université de Tours, 37380 Nouzilly, France; (P.C.); (E.R.)
| | - Eric Reiter
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Français du Cheval et de l’Equitation (IFCE), Université de Tours, 37380 Nouzilly, France; (P.C.); (E.R.)
| | - Clara Lazzaretti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Vincenzo Rochira
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| | - Giulia Brigante
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| | - Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Français du Cheval et de l’Equitation (IFCE), Université de Tours, 37380 Nouzilly, France; (P.C.); (E.R.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
10
|
Abstract
The glycoprotein follicle-stimulating hormone (FSH) acts on gonadal target cells, hence regulating gametogenesis. The transduction of the hormone-induced signal is mediated by the FSH-specific G protein-coupled receptor (FSHR), of which the action relies on the interaction with a number of intracellular effectors. The stimulatory Gαs protein is a long-time known transducer of FSH signaling, mainly leading to intracellular cAMP increase and protein kinase A (PKA) activation, the latter acting as a master regulator of cell metabolism and sex steroid production. While in vivo data clearly demonstrate the relevance of PKA activation in mediating gametogenesis by triggering proliferative signals, some in vitro data suggest that pro-apoptotic pathways may be awakened as a "dark side" of cAMP/PKA-dependent steroidogenesis, in certain conditions. P38 mitogen-activated protein kinases (MAPK) are players of death signals in steroidogenic cells, involving downstream p53 and caspases. Although it could be hypothesized that pro-apoptotic signals, if relevant, may be required for regulating atresia of non-dominant ovarian follicles, they should be transient and counterbalanced by mitogenic signals upon FSHR interaction with opposing transducers, such as Gαi proteins and β-arrestins. These molecules modulate the steroidogenic pathway via extracellular-regulated kinases (ERK1/2), phosphatidylinositol-4,5-bisphosphate 3-kinases (PI3K)/protein kinase B (AKT), calcium signaling and other intracellular signaling effectors, resulting in a complex and dynamic signaling network characterizing sex- and stage-specific gamete maturation. Even if the FSH-mediated signaling network is not yet entirely deciphered, its full comprehension is of high physiological and clinical relevance due to the crucial role covered by the hormone in regulating human development and reproduction.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
- *Correspondence: Livio Casarini
| | - Pascale Crépieux
- PRC, UMR INRA0085, CNRS 7247, Centre INRA Val de Loire, Nouzilly, France
| |
Collapse
|
11
|
Ulloa-Aguirre A, Reiter E, Crépieux P. FSH Receptor Signaling: Complexity of Interactions and Signal Diversity. Endocrinology 2018; 159:3020-3035. [PMID: 29982321 DOI: 10.1210/en.2018-00452] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022]
Abstract
FSH is synthesized in the pituitary by gonadotrope cells. By binding to and interacting with its cognate receptor [FSH receptor (FSHR)] in the gonads, this gonadotropin plays a key role in the control of gonadal function and reproduction. Upon activation, the FSHR undergoes conformational changes leading to transduction of intracellular signals, including dissociation of G protein complexes into components and activation of several associated interacting partners, which concertedly regulate downstream effectors. The canonical Gs/cAMP/protein kinase A pathway, considered for a long time as the sole effector of FSHR-mediated signaling, is now viewed as one of several mechanisms employed by this receptor to transduce intracellular signals in response to the FSH stimulus. This complex network of signaling pathways allows for a fine-tuning regulation of the gonadotropic stimulus, where activation/inhibition of its multiple components vary depending on the cell context, cell developmental stage, and concentration of associated receptors and corresponding ligands. Activation of these multiple signaling modules eventually converge to the hormone-integrated biological response, including survival, proliferation and differentiation of target cells, synthesis and secretion of paracrine/autocrine regulators, and, at the molecular level, functional selectivity and differential gene expression. In this mini-review, we discuss the complexity of FSHR-mediated intracellular signals activated in response to ligand stimulation. A better understanding of the signaling pathways involved in FSH action might potentially influence the development of new therapeutic strategies for reproductive disorders.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Eric Reiter
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| | - Pascale Crépieux
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| |
Collapse
|
12
|
Follicle-Stimulating Hormone Receptor: Advances and Remaining Challenges. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:1-58. [DOI: 10.1016/bs.ircmb.2018.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
13
|
Huang P, Zhou Z, Shi F, Shao G, Wang R, Wang J, Wang K, Ding W. Effects of the IGF-1/PTEN/Akt/FoxO signaling pathway on male reproduction in rats subjected to water immersion and restraint stress. Mol Med Rep 2016; 14:5116-5124. [PMID: 27779666 PMCID: PMC5355674 DOI: 10.3892/mmr.2016.5880] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 10/03/2016] [Indexed: 12/20/2022] Open
Abstract
The aim of the present study was to determine the effects of the insulin-like growth factor 1 (IGF-1)/phosphatase and tensin homologue deleted on chromosome 10 (PTEN)/Akt/forkhead box (FoxO) signaling pathway on male reproduction in rats subjected to water immersion and restraint stress (WRS). Sperm morphology, sperm malformation rate, and serum testosterone concentration were analyzed following WRS. In addition, the expression levels and immunolocalization of IGF-1, PTEN, Akt and FoxO proteins, as well as the rate of cell apoptosis in rat testes, were investigated. The results indicated that sperm malformation rate, serum testosterone concentration, and the number of terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells were increased in the testes after WRS. Furthermore, IGF-1 and FoxO1 proteins were predominantly localized in the sperm cytoplasm during the late stages of spermatogenesis. FoxO1 protein was also localized in Leydig cell cytoplasm. PTEN and total Akt proteins were predominantly expressed in the cytoplasm of Leydig cells and spermatogonia. PTEN protein was also detected in vascular endothelial cells. In addition, IGF-1, PTEN, Akt1, Akt2, FoxO3 and FoxO4 gene expression levels were upregulated following WRS, and peaked after 7 h of WRS. During the recovery period, the expression levels of these genes gradually returned to normal levels. The present study demonstrated that WRS induced sperm damage in the testes. In addition, the results indicated that the IGF-1/PTEN/Akt/FoxO signaling pathway may serve an anti-stress role in the testes of rats subjected to WRS.
Collapse
Affiliation(s)
- Pan Huang
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhengrong Zhou
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Fangxiong Shi
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Genbao Shao
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Ran Wang
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jintian Wang
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Kangxin Wang
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wei Ding
- Department of Animal Husbandry and Veterinary Medicine, Jiangsu Polytechnic College of Agriculture and Forestry, Jurong, Jiangsu 212400, P.R. China
| |
Collapse
|
14
|
Nascimento AR, Macheroni C, Lucas TFG, Porto CS, Lazari MFM. Crosstalk between FSH and relaxin at the end of the proliferative stage of rat Sertoli cells. Reproduction 2016; 152:613-628. [PMID: 27601715 DOI: 10.1530/rep-16-0330] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 09/05/2016] [Indexed: 12/31/2022]
Abstract
Follicle-stimulating hormone (FSH) stimulates the proliferation of immature Sertoli cells through the activation of PI3K/AKT/mTORC1 and MEK/ERK1/2 pathways. Mature Sertoli cells stop proliferating and respond to FSH by stimulating cAMP production. To gain insight into possible mechanisms involved in this switch as well as the impact of paracrine factors that stimulate cell proliferation, we analyzed the effects of FSH and relaxin on intracellular signaling pathways involved with proliferation and differentiation in Sertoli cells from 15-day-old rats, which are close to the transition between the two stages. FSH stimulated 3H-thymidine incorporation and cyclin D1 expression, changes associated with proliferation. In contrast, FSH inhibited AKT and ERK1/2 phosphorylation, activated cAMP production and induced changes in several cell cycle genes that were compatible with differentiation. Relaxin also stimulated 3H-thymidine incorporation but increased phosphorylation of ERK1/2 and AKT. When both hormones were added simultaneously, relaxin attenuated FSH-mediated inhibition of ERK1/2 and AKT phosphorylation and FSH-mediated activation of cAMP production. FSH but not relaxin increased CREB phosphorylation, and relaxin but not FSH shifted NF-κB expression from the cytoplasm to the nucleus. Relaxin did not inhibit the effects of FSH on inhibin α and Bcl2 expression. We propose that at this time of Sertoli cell development, FSH starts to direct cells to differentiation through activation of cAMP/CREB and inhibition of ERK1/2 and AKT pathways. Relaxin counteracts FSH signaling through the inhibition of cAMP and activation of ERK1/2, AKT and NF-κB, but does not block the differentiation process triggered by FSH.
Collapse
Affiliation(s)
- Aline R Nascimento
- Section of Experimental EndocrinologyDepartment of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Carla Macheroni
- Section of Experimental EndocrinologyDepartment of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Thais F G Lucas
- Section of Experimental EndocrinologyDepartment of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Catarina S Porto
- Section of Experimental EndocrinologyDepartment of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Maria F M Lazari
- Section of Experimental EndocrinologyDepartment of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| |
Collapse
|
15
|
Guillermet-Guibert J, Smith LB, Halet G, Whitehead MA, Pearce W, Rebourcet D, León K, Crépieux P, Nock G, Strömstedt M, Enerback M, Chelala C, Graupera M, Carroll J, Cosulich S, Saunders PTK, Huhtaniemi I, Vanhaesebroeck B. Novel Role for p110β PI 3-Kinase in Male Fertility through Regulation of Androgen Receptor Activity in Sertoli Cells. PLoS Genet 2015; 11:e1005304. [PMID: 26132308 PMCID: PMC4488938 DOI: 10.1371/journal.pgen.1005304] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 05/28/2015] [Indexed: 01/09/2023] Open
Abstract
The organismal roles of the ubiquitously expressed class I PI3K isoform p110β remain largely unknown. Using a new kinase-dead knockin mouse model that mimics constitutive pharmacological inactivation of p110β, we document that full inactivation of p110β leads to embryonic lethality in a substantial fraction of mice. Interestingly, the homozygous p110β kinase-dead mice that survive into adulthood (maximum ~26% on a mixed genetic background) have no apparent phenotypes, other than subfertility in females and complete infertility in males. Systemic inhibition of p110β results in a highly specific blockade in the maturation of spermatogonia to spermatocytes. p110β was previously suggested to signal downstream of the c-kit tyrosine kinase receptor in germ cells to regulate their proliferation and survival. We now report that p110β also plays a germ cell-extrinsic role in the Sertoli cells (SCs) that support the developing sperm, with p110β inactivation dampening expression of the SC-specific Androgen Receptor (AR) target gene Rhox5, a homeobox gene critical for spermatogenesis. All extragonadal androgen-dependent functions remain unaffected by global p110β inactivation. In line with a crucial role for p110β in SCs, selective inactivation of p110β in these cells results in male infertility. Our study is the first documentation of the involvement of a signalling enzyme, PI3K, in the regulation of AR activity during spermatogenesis. This developmental pathway may become active in prostate cancer where p110β and AR have previously been reported to functionally interact.
Collapse
Affiliation(s)
- Julie Guillermet-Guibert
- UCL Cancer Institute, University College London, London, United Kingdom
- Centre de Recherche en Cancérologie de Toulouse UMR1037, INSERM, BP84225, Toulouse, France
- Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Lee B. Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Guillaume Halet
- CNRS, UMR 6290, Institut de Génétique et Développement de Rennes, Rennes, France
- Université Rennes 1, UEB, SFR BIOSIT UMS 3480, Faculté de Médecine, Rennes, France
| | | | - Wayne Pearce
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Diane Rebourcet
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Kelly León
- Physiologie de la Reproduction et des Comportements, UMR 7247 INRA—CNRS—Université de Tours, Nouzilly, France
| | - Pascale Crépieux
- Physiologie de la Reproduction et des Comportements, UMR 7247 INRA—CNRS—Université de Tours, Nouzilly, France
| | - Gemma Nock
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Maria Strömstedt
- Astra Zeneca, Research and Development, Mölndal, Mölndal, Sweden
| | - Malin Enerback
- Astra Zeneca, Research and Development, Mölndal, Mölndal, Sweden
| | - Claude Chelala
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Mariona Graupera
- UCL Cancer Institute, University College London, London, United Kingdom
- Vascular Signalling Laboratory, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l’Hospitalet 199–203, 08908 L´Hospitalet de Llobregat, Barcelona, Spain
| | - John Carroll
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Sabina Cosulich
- Astrazeneca Oncology iMED, Alderley Park, Macclesfield, Cheshire, United Kingdom
| | - Philippa T. K. Saunders
- MRC Centre for Inflammation Research, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Ilpo Huhtaniemi
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, London, United Kingdom, and Department of Physiology, University of Turku, Turku, Finland
| | | |
Collapse
|
16
|
Sun X, Fu X, Li J, Xing C, Frierson HF, Wu H, Ding X, Ju T, Cummings RD, Dong JT. Deletion of atbf1/zfhx3 in mouse prostate causes neoplastic lesions, likely by attenuation of membrane and secretory proteins and multiple signaling pathways. Neoplasia 2014; 16:377-89. [PMID: 24934715 PMCID: PMC4198693 DOI: 10.1016/j.neo.2014.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/30/2014] [Accepted: 05/06/2014] [Indexed: 01/14/2023] Open
Abstract
The ATBF1/ZFHX3 gene at 16q22 is the second most frequently mutated gene in human prostate cancer and has reduced expression or mislocalization in several types of human tumors. Nonetheless, the hypothesis that ATBF1 has a tumor suppressor function in prostate cancer has not been tested. In this study, we examined the role of ATBF1 in prostatic carcinogenesis by specifically deleting Atbf1 in mouse prostatic epithelial cells. We also examined the effect of Atbf1 deletion on gene expression and signaling pathways in mouse prostates. Histopathologic analyses showed that Atbf1 deficiency caused hyperplasia and mouse prostatic intraepithelial neoplasia (mPIN) primarily in the dorsal prostate but also in other lobes. Hemizygous deletion of Atbf1 also increased the development of hyperplasia and mPIN, indicating a haploinsufficiency of Atbf1. The mPIN lesions expressed luminal cell markers and harbored molecular changes similar to those in human PIN and prostate cancer, including weaker expression of basal cell marker cytokeratin 5 (Ck5), cell adhesion protein E-cadherin, and the smooth muscle layer marker Sma; elevated expression of the oncoproteins phospho-Erk1/2, phospho-Akt and Muc1; and aberrant protein glycosylation. Gene expression profiling revealed a large number of genes that were dysregulated by Atbf1 deletion, particularly those that encode for secretory and cell membrane proteins. The four signaling networks that were most affected by Atbf1 deletion included those centered on Erk1/2 and IGF1, Akt and FSH, NF-κB and progesterone and β-estradiol. These findings provide in vivo evidence that ATBF1 is a tumor suppressor in the prostate, suggest that loss of Atbf1 contributes to tumorigenesis by dysregulating membrane and secretory proteins and multiple signaling pathways, and provide a new animal model for prostate cancer.
Collapse
Affiliation(s)
- Xiaodong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322
| | - Xiaoying Fu
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322; Department of Pathology, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jie Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322
| | - Changsheng Xing
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322
| | - Henry F Frierson
- Department of Pathology, University of Virginia Health System, Charlottesville, VA
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322
| | - Xiaokun Ding
- Department of Biochemistry, Emory University, Atlanta, GA 30322
| | - Tongzhong Ju
- Department of Biochemistry, Emory University, Atlanta, GA 30322
| | | | - Jin-Tang Dong
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA 30322.
| |
Collapse
|
17
|
Landomiel F, Gallay N, Jégot G, Tranchant T, Durand G, Bourquard T, Crépieux P, Poupon A, Reiter E. Biased signalling in follicle stimulating hormone action. Mol Cell Endocrinol 2014; 382:452-459. [PMID: 24121199 DOI: 10.1016/j.mce.2013.09.035] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/26/2013] [Accepted: 09/26/2013] [Indexed: 12/22/2022]
Abstract
Follicle-stimulating hormone (FSH) plays a crucial role in the control of reproduction by specifically binding to and activating a membrane receptor (FSHR) that belongs to the G protein-coupled receptor (GPCR) family. Similar to all GPCRs, FSHR activation mechanisms have generally been viewed as a two-state process connecting a unique FSH-bound active receptor to the Gs/cAMP pathway. Over the last decade, paralleling the breakthroughs that were made in the GPCR field, our understanding of FSH actions at the molecular level has dramatically changed. There are numerous facts indicating that the active FSHR is connected to a complex signalling network rather than the sole Gs/cAMP pathway. Consistently, the FSHR probably exists in equilibrium between multiple conformers, a subset of them being stabilized upon ligand binding. Importantly, the nature of the stabilized conformers of the receptor directly depends on the chemical structure of the ligand bound. This implies that it is possible to selectively control the intracellular signalling pathways activated by using biased ligands. Such biased ligands can be of different nature: small chemical molecules, glycosylation variants of the hormone or antibody/hormone complexes. Likewise, mutations or polymorphisms affecting the FSHR can also lead to stabilization of preferential conformers, hence to selective modulation of signalling pathways. These emerging notions offer a new conceptual framework that could potentially lead to the development of more specific drugs while also improving the way FSHR mutants/variants are functionally characterized.
Collapse
Affiliation(s)
- Flavie Landomiel
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Nathalie Gallay
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Gwenhael Jégot
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Thibaud Tranchant
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Guillaume Durand
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Thomas Bourquard
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Pascale Crépieux
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Anne Poupon
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Eric Reiter
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France.
| |
Collapse
|
18
|
Rocha BR, Colli SDR, Barcelos LM, Gregório BM, Sampaio FJB. Age-dependent expression of Pten and Smad4 genes in the urogenital system of Wistar rats. Acta Cir Bras 2014; 29 Suppl 1:34-8. [DOI: 10.1590/s0102-86502014001300007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
19
|
León K, Gallay N, Poupon A, Reiter E, Dalbies-Tran R, Crepieux P. Integrating microRNAs into the complexity of gonadotropin signaling networks. Front Cell Dev Biol 2013; 1:3. [PMID: 25364708 PMCID: PMC4206998 DOI: 10.3389/fcell.2013.00003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/12/2013] [Indexed: 12/13/2022] Open
Abstract
Follicle-stimulating hormone (FSH) is a master endocrine regulator of mammalian reproductive functions. Hence, it is used to stimulate folliculogenesis in assisted reproductive technologies (ART), both in women and in breeding animals. However, the side effects that hormone administration induces in some instances jeopardize the success of ART. Similarly, the luteinizing hormone (LH) is also of paramount importance in the reproductive function because it regulates steroidogenesis and the LH surge is a pre-requisite to ovulation. Gaining knowledge as extensive as possible on gonadotropin-induced biological responses could certainly lead to precise selection of their effects in vivo by the use of selective agonists at the hormone receptors. Hence, over the years, numerous groups have contributed to decipher the cellular events induced by FSH and LH in their gonadal target cells. Although little is known on the effect of gonadotropins on microRNA expression so far, recent data have highlighted that a microRNA regulatory network is likely to superimpose on the signaling protein network. No doubt that this will dramatically alter our current understanding of the gonadotropin-induced signaling networks. This is the topic of this review to present this additional level of complexity within the gonadotropin signaling network, in the context of recent findings on the microRNA machinery in the gonad.
Collapse
Affiliation(s)
- Kelly León
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Nathalie Gallay
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Anne Poupon
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Eric Reiter
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Rozenn Dalbies-Tran
- BINGO Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Pascale Crepieux
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| |
Collapse
|
20
|
Tarulli GA, Stanton PG, Meachem SJ. Is the adult Sertoli cell terminally differentiated? Biol Reprod 2012; 87:13, 1-11. [PMID: 22492971 DOI: 10.1095/biolreprod.111.095091] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
New data have challenged the convention that the adult Sertoli cell population is fixed and unmodifiable. The Sertoli cell has two distinct functions: 1) formation of the seminiferous cords and 2) provision of nutritional and structural support to developing germ cells. For these to occur successfully, Sertoli cells must undergo many maturational changes between fetal and adult life, the main switches occurring around puberty, including the loss of proliferative activity and the formation of the blood-testis barrier. Follicle-stimulating hormone plays a key role in promoting Sertoli cell proliferation, while thyroid hormone inhibits proliferative activity in early postnatal life. Together these regulate the Sertoli-germ cell complement and sperm output in adulthood. By puberty, the Sertoli cell population is considered to be stable and unmodifiable by hormones. But there is mounting evidence that the size of the adult Sertoli cell population and its maturational status is modifiable by hormones and that Sertoli cells can gain proliferative ability in the spermatogenically disrupted hamster and human model. This new information demonstrates that the adult Sertoli cell population, at least in the settings of testicular regression in the hamster and impaired fertility in humans in vivo and from mice and men in vitro, is not a terminally differentiated population. Data from the hamster now show that the adult Sertoli cell population size is regulated by hormones. This creates exciting prospects for basic and clinical research in testis biology. The potential to replenish an adult Sertoli-germ cell complement to normal in a setting of infertility may now be realized.
Collapse
Affiliation(s)
- Gerard A Tarulli
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| | | | | |
Collapse
|
21
|
Huang YS, Chen YM, Liao PC, Lee YH, Gwo JC, Chen MC, Chang CF. Testosterone improves the transition of primary oocytes in artificial maturation eels (Anguilla japonica) by altering ovarian PTEN expression. FISH PHYSIOLOGY AND BIOCHEMISTRY 2012; 38:777-787. [PMID: 21986810 DOI: 10.1007/s10695-011-9560-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 09/21/2011] [Indexed: 05/31/2023]
Abstract
In mammals, androgens appear to enhance the development of primary ovarian follicles, but PI3K (phosphoinositide 3-kinases) pathway is well recognized as one of the critical pathways in early follicular development. Roles of the PI3K were revealed by deletion of PTEN (phosphatase and tensin homolog on chromosome 10). PTEN is demonstrated to play an important role in the early stage of follicle development. In the Japanese eel, two forms of PTEN have been cloned, but what their functions on the development of early ovarian follicles are still not clear. The natural blockage and inducible of ovarian development was a benefit to address this question in the eel. Testosterone (T) shows to ameliorate the early ovarian development in the eel. The aims of this study were to elucidate the two forms of PTEN by cellular and physiological criteria and to study the effects of T on the ovarian PTEN production in the exogenous pituitary extracts-stimulated eel. Our results suggested that two forms of PTEN are existing in the Japanese eel, and eel ovarian development corresponded to the decrease in ovarian PTEN expression, vice versa. In addition, the supplement of T on eel early ovarian development can be attributed to its PTEN inhibitor role.
Collapse
Affiliation(s)
- Yung-Sen Huang
- Department of Life Science, National University of Kaohsiung, No. 700 Kaohsiung University Road, Nan Tzu Dist., Kaohsiung, Taiwan.
| | | | | | | | | | | | | |
Collapse
|
22
|
Zhang P, Chen JH, Guo XL. New insights into PTEN regulation mechanisms and its potential function in targeted therapies. Biomed Pharmacother 2012; 66:485-90. [PMID: 22902055 DOI: 10.1016/j.biopha.2012.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 04/15/2012] [Indexed: 12/12/2022] Open
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor gene of phosphatased activity. Its low or lacking expression closely relates with tumor progress and poor prognosis. The regulation and function ascribed to PTEN have become more diverse since its discovery as a putative phosphatase mutated in many human tumors. PTEN function is positively and negatively regulated at the transcriptional level, as well as post-translationally by phosphorylation, oxidation and acetylation. Deregulation of PTEN is implicated in other human diseases in addition to cancers, including diabetes and obesity, modulation of PTEN level has widespread therapeutic applications to those tumorigenesis and non-tumor diseases. This review will summarize the new points on the regulation of PTEN and briefly discuss the potential therapeutic role of PTEN in some diseases.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | | | | |
Collapse
|
23
|
Riera MF, Regueira M, Galardo MN, Pellizzari EH, Meroni SB, Cigorraga SB. Signal transduction pathways in FSH regulation of rat Sertoli cell proliferation. Am J Physiol Endocrinol Metab 2012; 302:E914-23. [PMID: 22275758 DOI: 10.1152/ajpendo.00477.2011] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The final number of Sertoli cells reached during the proliferative periods determines sperm production capacity in adulthood. It is well known that FSH is the major Sertoli cell mitogen; however, little is known about the signal transduction pathways that regulate the proliferation of Sertoli cells. The hypothesis of this investigation was that FSH regulates proliferation through a PI3K/Akt/mTORC1 pathway, and additionally, AMPK-dependent mechanisms counteract FSH proliferative effects. The present study was performed in 8-day-old rat Sertoli cell cultures. The results presented herein show that FSH, in addition to increasing p-Akt, p-mTOR, and p-p70S6K levels, increases p-PRAS40 levels, probably contributing to improving mTORC1 signaling. Furthermore, the decrease in FSH-stimulated p-Akt, p-mTOR, p-p70S6K, and p-PRAS40 levels in the presence of wortmannin emphasizes the participation of PI3K in FSH signaling. Additionally, the inhibition of FSH-stimulated Sertoli cell proliferation by the effect of wortmannin and rapamycin point to the relevance of the PI3K/Akt/mTORC1 signaling pathway in the mitotic activity of FSH. On the other hand, by activating AMPK, several interesting observations were made. Activation of AMPK produced an increase in Raptor phosphorylation, a decrease in p70S6K phosphorylation, and a decrease in FSH-stimulated Sertoli cell proliferation. The decrease in FSH-stimulated cell proliferation was accompanied by an increased expression of the cyclin-dependent kinase inhibitors (CDKIs) p19INK4d, p21Cip1, and p27Kip1. In summary, it is concluded that FSH regulates Sertoli cell proliferation with the participation of a PI3K/Akt/mTORC1 pathway and that AMPK activation may be involved in the detention of proliferation by, at least in part, a decrease in mTORC1 signaling and an increase in CDKI expression.
Collapse
Affiliation(s)
- María F Riera
- Centro de Investigaciones Endocrinológicas (CEDIE-CONICET), Hospital de Niños "R. Gutiérrez," Gallo, Buenos Aires, Argentina.
| | | | | | | | | | | |
Collapse
|
24
|
Musnier A, León K, Morales J, Reiter E, Boulo T, Costache V, Vourc'h P, Heitzler D, Oulhen N, Poupon A, Boulben S, Cormier P, Crépieux P. mRNA-selective translation induced by FSH in primary Sertoli cells. Mol Endocrinol 2012; 26:669-80. [PMID: 22383463 DOI: 10.1210/me.2011-1267] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
FSH is a key hormonal regulator of Sertoli cell secretory activity, required to optimize sperm production. To fulfil its biological function, FSH binds a G protein-coupled receptor, the FSH-R. The FSH-R-transduced signaling network ultimately leads to the transcription or down-regulation of numerous genes. In addition, recent evidence has suggested that FSH might also regulate protein translation. However, this point has never been demonstrated conclusively yet. Here we have addressed this issue in primary rat Sertoli cells endogenously expressing physiological levels of FSH-R. We observed that, within 90 min of stimulation, FSH not only enhanced overall protein synthesis in a mammalian target of rapamycin-dependent manner but also increased the recruitment of mRNA to polysomes. m(7)GTP pull-down experiments revealed the functional recruitment of mammalian target of rapamycin and p70 S6 kinase to the 5'cap, further supported by the enhanced phosphorylation of one of p70 S6 kinase targets, the eukaryotic initiation factor 4B. Importantly, the scaffolding eukaryotic initiation factor 4G was also recruited, whereas eukaryotic initiation factor 4E-binding protein, the eukaryotic initiation factor 4E generic inhibitor, appeared to play a minor role in translational regulations induced by FSH, in contrast to what is generally observed in response to anabolic factors. This particular regulation of the translational machinery by FSH stimulation might support mRNA-selective translation, as shown here by quantitative RT-PCR amplification of the c-fos and vascular endothelial growth factor mRNA but not of all FSH target mRNA, in polysomal fractions. These findings add a new level of complexity to FSH biological roles in its natural target cells, which has been underappreciated so far.
Collapse
Affiliation(s)
- Astrid Musnier
- BIOS Group, Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85, F-37380 Nouzilly, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lima-Fernandes E, Enslen H, Camand E, Kotelevets L, Boularan C, Achour L, Benmerah A, Gibson LCD, Baillie GS, Pitcher JA, Chastre E, Etienne-Manneville S, Marullo S, Scott MGH. Distinct functional outputs of PTEN signalling are controlled by dynamic association with β-arrestins. EMBO J 2011; 30:2557-68. [PMID: 21642958 DOI: 10.1038/emboj.2011.178] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 05/09/2011] [Indexed: 01/14/2023] Open
Abstract
The tumour suppressor PTEN (phosphatase and tensin deleted on chromosome 10) regulates major cellular functions via lipid phosphatase-dependent and -independent mechanisms. Despite its fundamental pathophysiological importance, how PTEN's cellular activity is regulated has only been partially elucidated. We report that the scaffolding proteins β-arrestins (β-arrs) are important regulators of PTEN. Downstream of receptor-activated RhoA/ROCK signalling, β-arrs activate the lipid phosphatase activity of PTEN to negatively regulate Akt and cell proliferation. In contrast, following wound-induced RhoA activation, β-arrs inhibit the lipid phosphatase-independent anti-migratory effects of PTEN. β-arrs can thus differentially control distinct functional outputs of PTEN important for cell proliferation and migration.
Collapse
|
26
|
Nicholls PK, Harrison CA, Walton KL, McLachlan RI, O'Donnell L, Stanton PG. Hormonal regulation of sertoli cell micro-RNAs at spermiation. Endocrinology 2011; 152:1670-83. [PMID: 21325043 DOI: 10.1210/en.2010-1341] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Spermatogenesis is absolutely dependent on FSH and androgens; suppression of these hormones inhibits germ cell development and thus sperm production. The final release of spermatids by the Sertoli cell, a process known as spermiation, is particularly sensitive to hormone suppression. To define the molecular mechanisms that mediate FSH and androgen effects in the Sertoli cell, we investigated the expression and regulation of micro-RNAs (miRNAs), small noncoding RNAs that regulate protein translation and modify cellular responses. By array analysis, we identified 23 miRNAs up-regulated more than 2-fold after hormone suppression in vivo and in vitro in primary Sertoli cell cultures. The regulation of four of these miRNAs (miR-23b, -30c, -30d, and -690) was confirmed by quantitative RT-PCR. Bioinformatic analysis of potential targets of hormonally regulated miRNAs identified genes important for focal adhesion and regulation of the actin cytoskeleton, processes known to be intimately associated with adhesion of spermatids to Sertoli cells. Two of the identified genes, Pten, an intracellular phosphatase, and Eps15, a mediator of endocytosis, were down-regulated by the withdrawal of hormones in vivo and possess miR-23b target sites in their 3'-untranslated regions. Overexpression of miR-23b in vitro resulted in decreased translation of PTEN and EPS15 protein as assessed by Western blot and luciferase analysis. We conclude that FSH and androgens act on Sertoli cells in stage VIII to control the expression of miRNAs that operate in a coordinated manner to regulate cell adhesion pathways and male fertility and that miRNA transcription is a new paradigm in the hormone dependence of spermiation.
Collapse
Affiliation(s)
- Peter K Nicholls
- Prince Henry's Institute, Monash Medical Centre, Clayton 3152, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
27
|
O'Donnell L, Nicholls PK, O'Bryan MK, McLachlan RI, Stanton PG. Spermiation: The process of sperm release. SPERMATOGENESIS 2011; 1:14-35. [PMID: 21866274 DOI: 10.4161/spmg.1.1.14525] [Citation(s) in RCA: 251] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 12/16/2010] [Accepted: 12/17/2010] [Indexed: 02/06/2023]
Abstract
Spermiation is the process by which mature spermatids are released from Sertoli cells into the seminiferous tubule lumen prior to their passage to the epididymis. It takes place over several days at the apical edge of the seminiferous epithelium, and involves several discrete steps including remodelling of the spermatid head and cytoplasm, removal of specialized adhesion structures and the final disengagement of the spermatid from the Sertoli cell. Spermiation is accomplished by the co-ordinated interactions of various structures, cellular processes and adhesion complexes which make up the "spermiation machinery". This review addresses the morphological, ultrastructural and functional aspects of mammalian spermiation. The molecular composition of the spermiation machinery, its dynamic changes and regulatory factors are examined. The causes of spermiation failure and their impact on sperm morphology and function are assessed in an effort to understand how this process may contribute to sperm count suppression during contraception and to phenotypes of male infertility.
Collapse
Affiliation(s)
- Liza O'Donnell
- Prince Henry's Institute of Medical Research; Clayton, VIC Australia
| | | | | | | | | |
Collapse
|
28
|
Gloaguen P, Crépieux P, Heitzler D, Poupon A, Reiter E. Mapping the follicle-stimulating hormone-induced signaling networks. Front Endocrinol (Lausanne) 2011; 2:45. [PMID: 22666216 PMCID: PMC3364461 DOI: 10.3389/fendo.2011.00045] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 09/14/2011] [Indexed: 01/14/2023] Open
Abstract
Follicle-stimulating hormone (FSH) is a central regulator of male and female reproductive function. Over the last decade, there has been a growing perception of the complexity associated with FSH-induced cellular signaling. It is now clear that the canonical Gs/cAMP/PKA pathway is not the sole mechanism that must be considered in FSH biological actions. In parallel, consistent with the emerging concept of biased agonism, several examples of ligand-mediated selective signaling pathway activation by gonadotropin receptors have been reported. In this context, it is important to gain an integrative view of the signaling pathways induced by FSH and how they interconnect to form a network. In this review, we propose a first attempt at building topological maps of various pathways known to be involved in the FSH-induced signaling network. We discuss the multiple facets of FSH-induced signaling and how they converge to the hormone integrated biological response. Despite of their incompleteness, these maps of the FSH-induced signaling network represent a first step toward gaining a system-level comprehension of this hormone's actions, which may ultimately facilitate the discovery of novel regulatory processes and therapeutic strategies for infertility and non-steroidal contraception.
Collapse
Affiliation(s)
- Pauline Gloaguen
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
| | - Pascale Crépieux
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
| | - Domitille Heitzler
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
| | - Anne Poupon
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
| | - Eric Reiter
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
- *Correspondence: Eric Reiter, INRA UMR85, CNRS-Université François Rabelais UMR6175, 37380, Nouzilly, France. e-mail:
| |
Collapse
|
29
|
Musnier A, Blanchot B, Reiter E, Crépieux P. GPCR signalling to the translation machinery. Cell Signal 2009; 22:707-16. [PMID: 19887105 DOI: 10.1016/j.cellsig.2009.10.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 10/23/2009] [Indexed: 12/26/2022]
Abstract
G protein-coupled receptors (GPCRs) are involved in most physiological processes, many of them being engaged in fully differentiated cells. These receptors couple to transducers of their own, primarily G proteins and beta-arrestins, which launch intracellular signalling cascades. Some of these signalling events regulate the translational machinery to fine-tune general cell metabolism or to alter protein expression pattern. Though extensively documented for tyrosine kinase receptors, translational regulation by GPCRs is still poorly appreciated. The objective of this review paper is to address the following questions: i) is there a "GPCR signature" impacting on the translational machinery, and ultimately on the type of mRNA translated? ii) are the regulatory networks involved similar as those utilized by tyrosine kinase receptors? In particular, we will discuss the specific features of translational control mediated by GPCRs and highlight the intrinsic properties of GPCRs these mechanisms could rely on.
Collapse
Affiliation(s)
- Astrid Musnier
- BIOS group, INRA, UMR, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | | | | | | |
Collapse
|