1
|
Bohaczuk SC, Tonsfeldt KJ, Slaiwa TI, Dunn GA, Gillette DLM, Yeo SE, Shi C, Cassin J, Thackray VG, Mellon PL. A Point Mutation in an Otherwise Dispensable Upstream Fshb Enhancer Moderately Impairs Fertility in Female Mice. Endocrinology 2025; 166:bqaf073. [PMID: 40237337 PMCID: PMC12038155 DOI: 10.1210/endocr/bqaf073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/18/2025]
Abstract
Follicle-stimulating hormone (FSH) is necessary for fertility in both sexes as a regulator of gametogenesis and hormone synthesis. Humans with loss-of-function mutations within the gene encoding the FSH beta subunit (FSHB) are infertile. Similarly, female Fshb knock-out mice are infertile and fail to ovulate, and males are subfertile. We recently reported the discovery and characterization of an upstream enhancer of FSHB located 26 Kb upstream of the transcriptional start site in humans (-17 Kb in mouse) that also amplifies activin and gonadotropin-releasing hormone induction of FSHB. Notably, the upstream enhancer contains a polymorphic, fertility-associated site in humans, rs11031006 (G/A), and the minor allele (A) increased enhancer activity in vitro as compared to the major allele (G), likely by increasing the affinity of an SF1 binding element. To investigate the role of the novel enhancer and rs11031006 variant in vivo, we created mouse models to assess deletion of the upstream enhancer and the impact of the G>A point mutation at the rs11031006-equivalent base. A full characterization of the -17 Kb enhancer deletion model revealed no apparent differences in fertility or serum FSH/LH levels, nor did a larger deletion that also included an additional putative regulatory element. In contrast, female mice homozygous for the mutated A allele at the rs11031006-equivalent position had fewer litters over a 120-day fertility assay, abnormal estrous cycling at 10 months, and reduced pituitary Lhb transcript abundance. Overall, while the mouse -17 Kb Fshb enhancer is dispensable for fertility, the rs11031006-equivalent G>A mutation results in subfertility in females.
Collapse
Affiliation(s)
- Stephanie C Bohaczuk
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Karen J Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Theresa I Slaiwa
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Geneva A Dunn
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dominique L M Gillette
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seung E Yeo
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chengxian Shi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jessica Cassin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Varykina G Thackray
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
2
|
Miyazawa K, Itoh Y, Fu H, Miyazono K. Receptor-activated transcription factors and beyond: multiple modes of Smad2/3-dependent transmission of TGF-β signaling. J Biol Chem 2024; 300:107256. [PMID: 38569937 PMCID: PMC11063908 DOI: 10.1016/j.jbc.2024.107256] [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: 01/19/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine that is widely distributed throughout the body. Its receptor proteins, TGF-β type I and type II receptors, are also ubiquitously expressed. Therefore, the regulation of various signaling outputs in a context-dependent manner is a critical issue in this field. Smad proteins were originally identified as signal-activated transcription factors similar to signal transducer and activator of transcription proteins. Smads are activated by serine phosphorylation mediated by intrinsic receptor dual specificity kinases of the TGF-β family, indicating that Smads are receptor-restricted effector molecules downstream of ligands of the TGF-β family. Smad proteins have other functions in addition to transcriptional regulation, including post-transcriptional regulation of micro-RNA processing, pre-mRNA splicing, and m6A methylation. Recent technical advances have identified a novel landscape of Smad-dependent signal transduction, including regulation of mitochondrial function without involving regulation of gene expression. Therefore, Smad proteins are receptor-activated transcription factors and also act as intracellular signaling modulators with multiple modes of function. In this review, we discuss the role of Smad proteins as receptor-activated transcription factors and beyond. We also describe the functional differences between Smad2 and Smad3, two receptor-activated Smad proteins downstream of TGF-β, activin, myostatin, growth and differentiation factor (GDF) 11, and Nodal.
Collapse
Affiliation(s)
- Keiji Miyazawa
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.
| | - Yuka Itoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Hao Fu
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Laboratory for Cancer Invasion and Metastasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
3
|
Cen S, Qian X, Wu C, Xu X, Yang X. Efficacy and Clinical Significance of the Zuogui Pill on Premature Ovarian Failure via the GDF-9/Smad2 Pathway. Nutr Cancer 2023; 75:488-497. [PMID: 36194038 DOI: 10.1080/01635581.2022.2123531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2022]
Abstract
Our study aims to investigate the efficacy and clinical significance of the Zuogui pill (ZGP) on premature ovarian failure (POF) via the GDF-9/Smad2 pathway. Changes in clinical symptoms in the control group (treated with Femoston alone) and the treatment group (treated with ZGP combined with Femoston) were assessed before and after treatment. Sex hormone levels, serum inflammatory cytokine levels, and ultrasound parameters were measured before and after treatment. POF rat models were established using cyclophosphamide and the POF rats were treated with Femoston, or ZGP combined with Femoston. GDF-9 and Smad2 expression levels were determined by RT-qPCR. The follicle-stimulating hormone (FSH), luteinizing hormone (LH), interleukin (IL)-6, and IL-21 levels, and the pulsatility index (PI) and resistance index (RI) values were decreased, while the estradiol (E2) and anti-Mullerian hormone (AMH) levels, antral follicle count (AFC), ovarian volume (OV), mean ovarian diameter (MOD), and peak systolic velocity (PSV) values were increased in the treatment group compared to the control group. After treatment with ZGP combined with Femoston, GDF-9 and Smad2 expression in the ovarian tissues of POF rats increased. ZGP has a therapeutic effect on POF via modulation of the GDF-9/Smad2 pathway.
Collapse
Affiliation(s)
- Su Cen
- Department of Gynecology, Hangzhou Xiaoshan District Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Xiaojun Qian
- Department of Gynecology, Hangzhou Xiaoshan District Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Chunfang Wu
- Department of Traditional Chinese Medicine, Maternal and Child Health Care Hospital of Zhuji, Zhuji, Zhejiang, China
| | - Xinya Xu
- Gynaecologic Department of Traditional Chinese Medicine, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Xiaohui Yang
- Department of Gynecology, Hangzhou Xiaoshan District Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Bhattacharya I, Dey S, Banerjee A. Revisiting the gonadotropic regulation of mammalian spermatogenesis: evolving lessons during the past decade. Front Endocrinol (Lausanne) 2023; 14:1110572. [PMID: 37124741 PMCID: PMC10140312 DOI: 10.3389/fendo.2023.1110572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Spermatogenesis is a multi-step process of male germ cell (Gc) division and differentiation which occurs in the seminiferous tubules of the testes under the regulation of gonadotropins - Follicle Stimulating Hormone (FSH) and Luteinising hormone (LH). It is a highly coordinated event regulated by the surrounding somatic testicular cells such as the Sertoli cells (Sc), Leydig cells (Lc), and Peritubular myoid cells (PTc). FSH targets Sc and supports the expansion and differentiation of pre-meiotic Gc, whereas, LH operates via Lc to produce Testosterone (T), the testicular androgen. T acts on all somatic cells e.g.- Lc, PTc and Sc, and promotes the blood-testis barrier (BTB) formation, completion of Gc meiosis, and spermiation. Studies with hypophysectomised or chemically ablated animal models and hypogonadal (hpg) mice supplemented with gonadotropins to genetically manipulated mouse models have revealed the selective and synergistic role(s) of hormones in regulating male fertility. We here have briefly summarized the present concept of hormonal control of spermatogenesis in rodents and primates. We also have highlighted some of the key critical questions yet to be answered in the field of male reproductive health which might have potential implications for infertility and contraceptive research in the future.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, School of Biological Science, Central University of Kerala, Kasaragod, Kerala, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| | - Souvik Dey
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Arnab Banerjee
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Goa, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| |
Collapse
|
5
|
Lalonde-Larue A, Boyer A, Dos Santos EC, Boerboom D, Bernard DJ, Zamberlam G. The Hippo Pathway Effectors YAP and TAZ Regulate LH Release by Pituitary Gonadotrope Cells in Mice. Endocrinology 2022; 163:bqab238. [PMID: 34905605 PMCID: PMC8670590 DOI: 10.1210/endocr/bqab238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Indexed: 01/08/2023]
Abstract
The Hippo transcriptional coactivators YAP and TAZ exert critical roles in morphogenesis, organ size determination and tumorigenesis in many tissues. Although Hippo kinase cascade activity was recently reported in the anterior pituitary gland in mice, the role of the Hippo effectors in regulating gonadotropin production remains unknown. The objective of this study was therefore to characterize the roles of YAP and TAZ in gonadotropin synthesis and secretion. Using a conditional gene targeting approach (cKO), we found that gonadotrope-specific inactivation of Yap and Taz resulted in increased circulating levels of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) in adult male mice, along with increased testosterone levels and testis weight. Female cKO mice had increased circulating LH (but not FSH) levels, which were associated with a hyperfertility phenotype characterized by higher ovulation rates and larger litter sizes. Unexpectedly, the loss of YAP/TAZ did not appear to affect the expression of gonadotropin subunit genes, yet both basal and GnRH-induced LH secretion were increased in cultured pituitary cells from cKO mice. Likewise, pharmacologic inhibition of YAP binding to the TEAD family of transcription factors increased both basal and GnRH-induced LH secretion in LβT2 gonadotrope-like cells in vitro without affecting Lhb expression. Conversely, mRNA levels of ChgA and SgII, which encode key secretory granule cargo proteins, were decreased following pharmacologic inhibition of YAP/TAZ, suggesting a mechanism whereby YAP/TAZ regulate the LH secretion machinery in gonadotrope cells. Together, these findings represent the first evidence that Hippo signaling may play a role in regulating pituitary LH secretion.
Collapse
Affiliation(s)
- Ariane Lalonde-Larue
- Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine Vétérinaire, Université de Montréal, Montréal, Quebec J2S 7C6, Canada
| | - Alexandre Boyer
- Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine Vétérinaire, Université de Montréal, Montréal, Quebec J2S 7C6, Canada
| | - Esdras Corrêa Dos Santos
- Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine Vétérinaire, Université de Montréal, Montréal, Quebec J2S 7C6, Canada
| | - Derek Boerboom
- Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine Vétérinaire, Université de Montréal, Montréal, Quebec J2S 7C6, Canada
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Gustavo Zamberlam
- Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine Vétérinaire, Université de Montréal, Montréal, Quebec J2S 7C6, Canada
| |
Collapse
|
6
|
Yang H, Wan Z, Jin Y, Wang F, Zhang Y. SMAD2 regulates testicular development and testosterone synthesis in Hu sheep. Theriogenology 2021; 174:139-148. [PMID: 34454319 DOI: 10.1016/j.theriogenology.2021.08.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 08/15/2021] [Accepted: 08/22/2021] [Indexed: 01/01/2023]
Abstract
The SMAD protein family plays crucial roles in reproduction as a downstream target genes of the TGFβ signaling pathway. Many studies have focused on the expression change exploration of SMADs during testicular development and investigation of SMAD2 in hormone synthesis regulation. However, little attention has been given to determining the regulatory mechanism of SMADs in sheep testes. In the present study, we first detected SMAD mRNA expression levels in three-month-old (3 M), six-month-old (6 M), nine-month-old (9 M) and two-year-old (2Y) sheep testes. Different SMADs showed various expression patterns. In addition, the subcellular localization of SMAD2 was also analyzed, and Sertoli cells (SCs), Leydig cells (LCs) and spermatogonia presented mainly positive staining. Protein and nucleic acid sequence alignment showed that the SMAD2 gene was extremely homologous between various species. SMAD2 interference RNA was transfected into sheep LCs to examine the cell proliferation and hormone levels. The testosterone level was significantly decreased, and cell proliferation efficiency presented the same trend (P < 0.05). Moreover, SMAD2 downregulation promoted cell apoptosis (P < 0.05) and changed the cell cycle. In total, our results revealed that downregulating the expression of SMAD2 can effectively inhibit testosterone levels by affecting cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Hua Yang
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zhen Wan
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yanshan Jin
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Feng Wang
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yanli Zhang
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
7
|
Ongaro L, Alonso CAI, Zhou X, Brûlé E, Li Y, Schang G, Parlow AF, Steyn F, Bernard DJ. Development of a Highly Sensitive ELISA for Measurement of FSH in Serum, Plasma, and Whole Blood in Mice. Endocrinology 2021; 162:6105044. [PMID: 33475143 PMCID: PMC7894055 DOI: 10.1210/endocr/bqab014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2020] [Indexed: 01/09/2023]
Abstract
Follicle-stimulating hormone (FSH) regulates gonadal function and fertility. Measurement of FSH in bodily fluids and tissues is possible with radioimmunoassays and enzyme-linked immunosorbent assays (ELISAs). Recently, several novel assays were developed to measure pituitary hormones including growth hormone, prolactin, and luteinizing hormone in mice from small sample volumes. Here, we describe a novel and sensitive ELISA that enables the accurate measurement of FSH in serum, plasma, and whole blood from female and male mice. The assay can also be used to measure FSH in murine pituitary lysates and cell culture media. In summary, the new methodology described here will enable investigators to measure FSH from a variety of biological samples in mice accurately, at low cost, and in their own laboratories.
Collapse
Affiliation(s)
- Luisina Ongaro
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | | | - Xiang Zhou
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Emilie Brûlé
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Yining Li
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Gauthier Schang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Albert F Parlow
- National Hormone & Peptide Program, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Frederik Steyn
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Neurology, Royal Brisbane & Women’s Hospital, Queensland, Brisbane, Australia
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
- Correspondence: Daniel J. Bernard, PhD, McGill University, Montreal, QC, Canada.
| |
Collapse
|
8
|
Huo JF, Zhang ML, Wang XX, Zou DH. Chrysin induces osteogenic differentiation of human dental pulp stem cells. Exp Cell Res 2021; 400:112466. [PMID: 33508275 DOI: 10.1016/j.yexcr.2020.112466] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/20/2020] [Accepted: 12/27/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVES As an ideal cell source for tissue engineering and bone defect repair, dental pulp stem cells (DPSCs) have good osteogenic differentiation potential. Chrysin, a flavonoid extracted from oroxylum seeds, has been proven to promote bone formation of bone marrow stem cells. However, the effect of chrysin on osteogenic differentiation of DPSCs remains unclear. This study aimed to investigate the role of Chrysin in promoting osteogenic differentiation of DPSCs and in DPSC-based bone formation. MATERIAL AND METHODS We investigated the effects of chrysin on DPSCs from patients by CCK-8 assay, Alizarin Red S staining, qPCR and Western blotting. The effects of chrysin on DPSC-based bone formation in a heterotopic osteogenesis model in nude mice and a rat calvarial defect model were also performed. Finally, we investigated the mechanism of chrysin-treated DPSCs by proteomics. RESULTS Chrysin upregulated the expression of osteogenic proteins and induced osteogenic differentiation of DPSCs. Moreover, chrysin induced abundant β-TCP-induced formation of mineralized bone tissue and promoted DPSC-based bone formation in a heterotopic osteogenesis model in nude mice and a rat calvarial defect model. Proteomics showed that upregulation of the Smad3 was closely related to osteogenic differentiation. Inhibiting of Smad3 activation by a Smad3 inhibitor could reverse the chrysin-mediated increases in the expression levels of osteogenic genes and osteogenic induction of DPSCs. CONCLUSIONS Our study implies the intriguing potential of chrysin-treated DPSCs in bone regeneration and bone defect repair.
Collapse
Affiliation(s)
- J F Huo
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Department of Stomatology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - M L Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - X X Wang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.
| | - D H Zou
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China.
| |
Collapse
|
9
|
Kharazi U, Badalzadeh R. A review on the stem cell therapy and an introduction to exosomes as a new tool in reproductive medicine. Reprod Biol 2020; 20:447-459. [DOI: 10.1016/j.repbio.2020.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/18/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022]
|
10
|
Wang Y, Zhang X, Mao Y, Liang L, Liu L, Peng W, Liu H, Xiao Y, Zhang Y, Zhang F, Shi M, Liu L, Guo B. Smad2 and Smad3 play antagonistic roles in high glucose-induced renal tubular fibrosis via the regulation of SnoN. Exp Mol Pathol 2020; 113:104375. [PMID: 31917288 DOI: 10.1016/j.yexmp.2020.104375] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/10/2019] [Accepted: 01/04/2020] [Indexed: 12/22/2022]
Abstract
Diabetic nephropathy (DN) is a serious microvascular complication of diabetes mellitus.The main pathological features of DN include glomerular sclerosis and renal tubular interstitial fibrosis, which results in epithelial mesenchymal transition (EMT) and excessive extracellular matrix (ECM) deposition.Transforming growth factor-β1(TGF-β1) is a critical factor that regulates the manifestation of renal fibrosis.Smad2 and Smad3 are the main downstream of the TGF-β1 pathway. Ski-related novel protein N(SnoN) is a negative regulator of TGF-β1, and inhibits the activation of the TGF-β1/Smad2/3 signalling pathway. In this study, the expression of Smad2 and Smad3 proteins, SnoN mRNA, SnoN proteins, and the ubiquitination levels of SnoN were determined in DN rats and renal tubular epithelial cells(NRK52E cells). Knockdown and overexpression of Smad2 or Smad3 in NRK52E cells were used to investigate the specific roles of Smad2 and Smad3 in the development of high glucose-induced renal tubular fibrosis, with a specific focus on their effect on the regulation of SnoN expression. Our study demonstrated that Smad3 could inhibit SnoN expression and increase ECM deposition in NRK52E cells, to promote high glucose-induced renal tubular fibrosis. In contrast, Smad2 could induce SnoN expression and reduce ECM deposition, to inhibit high glucose-induced fibrosis. The underlying mechanism involves regulation of SnoN expression. These findings provide a novel mechanism to understanding the significant role of the TGF-β1/ Smad2/3 pathway in DN.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiaohuan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yanwen Mao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Luqun Liang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Lingling Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Wei Peng
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Huiming Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yingying Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Lirong Liu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China; Department of Clinical Hematology, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
11
|
Li Y, Fortin J, Ongaro L, Zhou X, Boehm U, Schneyer A, Bernard DJ, Lin HY. Betaglycan (TGFBR3) Functions as an Inhibin A, but Not Inhibin B, Coreceptor in Pituitary Gonadotrope Cells in Mice. Endocrinology 2018; 159:4077-4091. [PMID: 30364975 PMCID: PMC6372943 DOI: 10.1210/en.2018-00770] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/22/2018] [Indexed: 02/01/2023]
Abstract
Inhibins are gonadal hormones that act on pituitary gonadotrope cells to suppress FSH synthesis and secretion. Inhibin A and B are heterodimers of the inhibin ⍺-subunit disulfide-linked to one of two inhibin β-subunits. Homodimers or heterodimers of the inhibin β-subunits form the activins, which stimulate FSH production. Activins signal through complexes of type I and II receptor serine/threonine kinases to increase transcription of the FSHβ subunit gene. According to in vitro observations, inhibins impair FSH synthesis by competitively binding to activin type II receptors, particularly in the presence of the TGFβ type III receptor (TGFBR3, or betaglycan). The role of TGFBR3 in inhibin action in vivo has not been determined. Here, we ablated Tgfbr3 specifically in murine gonadotropes. Conditional knockout females were supra-fertile, exhibiting enhanced folliculogenesis, numbers of ovulated eggs per cycle, and litter sizes relative to control mice. Despite these phenotypes, FSH levels appeared to be unaltered in knockout mice, and the mechanisms underlying their enhanced fertility remain unexplained. Inhibin B is the predominant form of the hormone in males and in females during most stages of the estrous cycle. Remarkably, inhibin A, but not inhibin B, suppression of FSH synthesis was impaired in cultured pituitaries of knockout mice, which may explain the absence of discernible changes in FSH levels in vivo. Collectively, these data challenge current dogma by demonstrating that TGFBR3 (betaglycan) functions as an inhibin A, but not an inhibin B, coreceptor in gonadotrope cells in vivo. Mechanisms of inhibin B action merit further investigation.
Collapse
Affiliation(s)
- Yining Li
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Jérôme Fortin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Luisina Ongaro
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Xiang Zhou
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology, University of Saarland School of Medicine, Homburg, Germany
| | | | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Herbert Y Lin
- Program in Membrane Biology/Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
12
|
Huang B, Lu J, Ding C, Zou Q, Wang W, Li H. Exosomes derived from human adipose mesenchymal stem cells improve ovary function of premature ovarian insufficiency by targeting SMAD. Stem Cell Res Ther 2018; 9:216. [PMID: 30092819 PMCID: PMC6085638 DOI: 10.1186/s13287-018-0953-7] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/26/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023] Open
Abstract
Background Although many reports show that various kinds of stem cells have the ability to recover the function of premature ovarian insufficiency (POI), few studies are associated with the mechanism of stem cell treatment of POI. We designed this experimental study to investigate whether human adipose stem cell-derived exosomes (hADSC-Exos) retain the ability to restore ovarian function and how hADSC-Exos work in this process. Methods A POI mouse model was established and human ovarian granule cells (hGCs) collected from individuals with POI were prepared to assess the therapeutic effects and illuminate the mechanism of hADSCs in curing POI. The hematoxylin and eosin assay method was employed to assess the number of follicles. Enzyme-linked immunosorbent assay (ELISA) was used to detect the serum levels of sex hormones. The proliferation rate and marker expression levels of hGCs were measured by flow cytometry (fluorescence-activated cell sorting). Real-time PCR and western blot assays were used to determine the mRNA and protein expression levels of SMAD2, SMAD3, and SMAD5. Western blot assays were used to test the protein expression levels of apoptosis genes (Fas, FasL, caspase-3, and caspase-8). Results After the hADSC-Exos were transplanted into the POI mice model, they exerted better therapeutic activity on mouse ovarian function, improving follicle numbers during four stages. ELISA results showed that hADSC-Exos elevated the hormone levels to the normal levels. In addition, after hADSC-Exos were cocultured with POI hGCs, our results showed that hADSC-Exos significantly promoted the proliferation rate and inhibited the apoptosis rate. Furthermore, hADSC-Exos also increased the marker expression of hGCs to the normal level. Besides, mRNA and protein assays demonstrated that hADSC-Exos downregulated the expression of SMAD2, SMAD3, and SMAD5 in vivo and in vitro. Western blot assay demonstrated that hADSC-Exos inhibited expression of the apoptosis genes in POI hGCs, and SMAD knockdown increased the protein expression of apoptosis genes. Conclusions These findings demonstrate for the first time the molecular cascade and related cell biology events involved in the mechanism by which exosomes derived from hADSCs improved ovarian function of POI disease via regulation of the SMAD signaling pathway. Electronic supplementary material The online version of this article (10.1186/s13287-018-0953-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Boxian Huang
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China. .,Central Laboratory, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China. .,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.
| | - Jiafeng Lu
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Chenyue Ding
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Qinyan Zou
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Wei Wang
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Hong Li
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China.
| |
Collapse
|
13
|
Rimon-Dahari N, Heinemann-Yerushalmi L, Hadas R, Kalich-Philosoph L, Ketter D, Nevo N, Galiani D, Dekel N. Vasorin: a newly identified regulator of ovarian folliculogenesis. FASEB J 2018; 32:2124-2136. [PMID: 29259033 DOI: 10.1096/fj.201700057rrr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Members of the TGF-β superfamily take part in the control of folliculogenesis. Vasorin (Vasn) is a newly identified negative regulator of TGF-β signaling whose possible involvement in ovarian physiology has never been studied. Here, we demonstrate that Vasn is expressed in the ovary by somatic cells of follicles, and that its expression is up-regulated by LH. We established a conditional knockout (cKO) mouse model in which Vasn is deleted specifically in granulosa cells of growing follicles from the secondary stage onwards. Using this model, we show that, upon hormonal stimulation, follicle ovulation size is almost 2-fold higher. This enhanced ovulatory response is associated with overactivation of the TGF-β signaling pathway and a lower number of atretic antral follicles. Of importance, we demonstrate that the number of primordial follicles is reduced in prepubertal cKO mouse ovaries, which suggests that the production of VASN by growing follicles protects the ovarian reserve. Finally, analysis of systemic KO mice revealed that the ovarian reserve is almost 2.5-fold higher, which implies that Vasn may also play a role in primordial follicle formation. Overall, our findings reveal that Vasn is a new regulator that exerts an effect on several key ovarian functions, including folliculogenesis, maintenance of the ovarian reserve, and ovulation.-Rimon-Dahari, N., Heinemann-Yerushalmi, L., Hadas, R., Kalich-Philosoph, L., Ketter, D., Nevo, N., Galiani, D., Dekel, N. Vasorin: a newly identified regulator of ovarian folliculogenesis.
Collapse
Affiliation(s)
- Nitzan Rimon-Dahari
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Ron Hadas
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Dafna Ketter
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Nava Nevo
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Dalia Galiani
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Nava Dekel
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
14
|
Coss D. Regulation of reproduction via tight control of gonadotropin hormone levels. Mol Cell Endocrinol 2018; 463:116-130. [PMID: 28342855 PMCID: PMC6457911 DOI: 10.1016/j.mce.2017.03.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 01/04/2023]
Abstract
Mammalian reproduction is controlled by the hypothalamic-pituitary-gonadal axis. GnRH from the hypothalamus regulates synthesis and secretion of gonadotropins, LH and FSH, which then control steroidogenesis and gametogenesis. In females, serum LH and FSH levels exhibit rhythmic changes throughout the menstrual or estrous cycle that are correlated with pulse frequency of GnRH. Lack of gonadotropins leads to infertility or amenorrhea. Dysfunctions in the tightly controlled ratio due to levels slightly outside the normal range occur in a larger number of women and are correlated with polycystic ovaries and premature ovarian failure. Since the etiology of these disorders is largely unknown, studies in cell and mouse models may provide novel candidates for investigations in human population. Hence, understanding the mechanisms whereby GnRH regulates gonadotropin hormone levels will provide insight into the physiology and pathophysiology of the reproductive system. This review discusses recent advances in our understanding of GnRH regulation of gonadotropin synthesis.
Collapse
Affiliation(s)
- Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA 92521, United States.
| |
Collapse
|
15
|
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
|
16
|
Jonak CR, Lainez NM, Boehm U, Coss D. GnRH Receptor Expression and Reproductive Function Depend on JUN in GnRH Receptor‒Expressing Cells. Endocrinology 2018; 159:1496-1510. [PMID: 29409045 PMCID: PMC5839737 DOI: 10.1210/en.2017-00844] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/10/2018] [Indexed: 12/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) from the hypothalamus regulates synthesis and secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) from the anterior pituitary gonadotropes. LH and FSH are heterodimers composed of a common α-subunit and unique β-subunits, which provide biological specificity and are limiting components of mature hormone synthesis. Gonadotrope cells respond to GnRH via specific expression of the GnRH receptor (Gnrhr). GnRH induces the expression of gonadotropin genes and of the Gnrhr by activation of specific transcription factors. The JUN (c-Jun) transcription factor binds to AP-1 sites in the promoters of target genes and mediates induction of the FSHβ gene and of the Gnrhr in gonadotrope-derived cell lines. To analyze the role of JUN in reproductive function in vivo, we generated a mouse model that lacks JUN specifically in GnRH receptor‒expressing cells (conditional JUN knockout; JUN-cKO). JUN-cKO mice displayed profound reproductive anomalies such as reduced LH levels resulting in lower gonadal steroid levels, longer estrous cycles in females, and diminished sperm numbers in males. Unexpectedly, FSH levels were unchanged in these animals, whereas Gnrhr expression in the pituitary was reduced. Steroidogenic enzyme expression was reduced in the gonads of JUN-cKO mice, likely as a consequence of reduced LH levels. GnRH receptor‒driven Cre activity was detected in the hypothalamus but not in the GnRH neuron. Female, but not male, JUN-cKO mice exhibited reduced GnRH expression. Taken together, our results demonstrate that GnRH receptor‒expression levels depend on JUN and are critical for reproductive function.
Collapse
Affiliation(s)
- Carrie R. Jonak
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Nancy M. Lainez
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, 66421 Homburg, Germany
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California 92521
- Correspondence: Djurdjica Coss, PhD, Division of Biomedical Sciences, School of Medicine, 303 SOM Research Building, University of California, Riverside, Riverside, California 92521. E-mail:
| |
Collapse
|
17
|
Odle AK, Akhter N, Syed MM, Allensworth-James ML, Beneš H, Melgar Castillo AI, MacNicol MC, MacNicol AM, Childs GV. Leptin Regulation of Gonadotrope Gonadotropin-Releasing Hormone Receptors As a Metabolic Checkpoint and Gateway to Reproductive Competence. Front Endocrinol (Lausanne) 2018; 8:367. [PMID: 29354094 PMCID: PMC5760501 DOI: 10.3389/fendo.2017.00367] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/13/2017] [Indexed: 12/20/2022] Open
Abstract
The adipokine leptin signals the body's nutritional status to the brain, and particularly, the hypothalamus. However, leptin receptors (LEPRs) can be found all throughout the body and brain, including the pituitary. It is known that leptin is permissive for reproduction, and mice that cannot produce leptin (Lep/Lep) are infertile. Many studies have pinpointed leptin's regulation of reproduction to the hypothalamus. However, LEPRs exist at all levels of the hypothalamic-pituitary-gonadal axis. We have previously shown that deleting the signaling portion of the LEPR specifically in gonadotropes impairs fertility in female mice. Our recent studies have targeted this regulation to the control of gonadotropin releasing hormone receptor (GnRHR) expression. The hypotheses presented here are twofold: (1) cyclic regulation of pituitary GnRHR levels sets up a target metabolic checkpoint for control of the reproductive axis and (2) multiple checkpoints are required for the metabolic signaling that regulates the reproductive axis. Here, we emphasize and explore the relationship between the hypothalamus and the pituitary with regard to the regulation of GnRHR. The original data we present strengthen these hypotheses and build on our previous studies. We show that we can cause infertility in 70% of female mice by deleting all isoforms of LEPR specifically in gonadotropes. Our findings implicate activin subunit (InhBa) mRNA as a potential leptin target in gonadotropes. We further show gonadotrope-specific upregulation of GnRHR protein (but not mRNA levels) following leptin stimulation. In order to try and understand this post-transcriptional regulation, we tested candidate miRNAs (identified with in silico analysis) that may be binding the Gnrhr mRNA. We show significant upregulation of one of these miRNAs in our gonadotrope-Lepr-null females. The evidence provided here, combined with our previous work, lay the foundation for metabolically regulated post-transcriptional control of the gonadotrope. We discuss possible mechanisms, including miRNA regulation and the involvement of the RNA binding protein, Musashi. We also demonstrate how this regulation may be vital for the dynamic remodeling of gonadotropes in the cycling female. Finally, we propose that the leptin receptivity of both the hypothalamus and the pituitary are vital for the body's ability to delay or slow reproduction during periods of low nutrition.
Collapse
Affiliation(s)
- Angela K. Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Noor Akhter
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Mohsin M. Syed
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Melody L. Allensworth-James
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Helen Beneš
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Andrea I. Melgar Castillo
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Melanie C. MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Angus M. MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Gwen V. Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
18
|
Monsivais D, Matzuk MM, Pangas SA. The TGF-β Family in the Reproductive Tract. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022251. [PMID: 28193725 DOI: 10.1101/cshperspect.a022251] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The transforming growth factor β (TGF-β) family has a profound impact on the reproductive function of various organisms. In this review, we discuss how highly conserved members of the TGF-β family influence the reproductive function across several species. We briefly discuss how TGF-β-related proteins balance germ-cell proliferation and differentiation as well as dauer entry and exit in Caenorhabditis elegans. In Drosophila melanogaster, TGF-β-related proteins maintain germ stem-cell identity and eggshell patterning. We then provide an in-depth analysis of landmark studies performed using transgenic mouse models and discuss how these data have uncovered basic developmental aspects of male and female reproductive development. In particular, we discuss the roles of the various TGF-β family ligands and receptors in primordial germ-cell development, sexual differentiation, and gonadal cell development. We also discuss how mutant mouse studies showed the contribution of TGF-β family signaling to embryonic and postnatal testis and ovarian development. We conclude the review by describing data obtained from human studies, which highlight the importance of the TGF-β family in normal female reproductive function during pregnancy and in various gynecologic pathologies.
Collapse
Affiliation(s)
- Diana Monsivais
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030
| | - Martin M Matzuk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030.,Department of Molecular and Cellular Biology, Baylor College of Medicine Houston, Texas 77030.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030.,Department of Pharmacology, Baylor College of Medicine, Houston, Texas 77030
| | - Stephanie A Pangas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030.,Department of Molecular and Cellular Biology, Baylor College of Medicine Houston, Texas 77030
| |
Collapse
|
19
|
Li Y, Schang G, Boehm U, Deng CX, Graff J, Bernard DJ. SMAD3 Regulates Follicle-stimulating Hormone Synthesis by Pituitary Gonadotrope Cells in Vivo. J Biol Chem 2016; 292:2301-2314. [PMID: 27994055 DOI: 10.1074/jbc.m116.759167] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
Pituitary follicle-stimulating hormone (FSH) is an essential regulator of fertility in females and of quantitatively normal spermatogenesis in males. Pituitary-derived activins are thought to act as major stimulators of FSH synthesis by gonadotrope cells. In vitro, activins signal via SMAD3, SMAD4, and forkhead box L2 (FOXL2) to regulate transcription of the FSHβ subunit gene (Fshb). Consistent with this model, gonadotrope-specific Smad4 or Foxl2 knock-out mice have greatly reduced FSH and are subfertile. The role of SMAD3 in vivo is unresolved; however, residual FSH production in Smad4 conditional knock-out mice may derive from partial compensation by SMAD3 and its ability to bind DNA in the absence of SMAD4. To test this hypothesis and determine the role of SMAD3 in FSH biosynthesis, we generated mice lacking both the SMAD3 DNA binding domain and SMAD4 specifically in gonadotropes. Conditional knock-out females were hypogonadal, acyclic, and sterile and had thread-like uteri; their ovaries lacked antral follicles and corpora lutea. Knock-out males were fertile but had reduced testis weights and epididymal sperm counts. These phenotypes were consistent with those of Fshb knock-out mice. Indeed, pituitary Fshb mRNA levels were nearly undetectable in both male and female knock-outs. In contrast, gonadotropin-releasing hormone receptor mRNA levels were significantly elevated in knock-outs in both sexes. Interestingly, luteinizing hormone production was altered in a sex-specific fashion. Overall, our analyses demonstrate that SMAD3 is required for FSH synthesis in vivo.
Collapse
Affiliation(s)
- Yining Li
- From the Centre for Research in Reproduction and Development, Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Gauthier Schang
- From the Centre for Research in Reproduction and Development, Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Ulrich Boehm
- the Department of Pharmacology and Toxicology, University of Saarland School of Medicine, D-66421 Homburg, Germany
| | - Chu-Xia Deng
- the Faculty of Health Sciences, University of Macau, Macau SAR 999078, China, and
| | - Jonathan Graff
- the Department of Developmental Biology, University of Texas Southwestern, Dallas, Texas 75390
| | - Daniel J Bernard
- From the Centre for Research in Reproduction and Development, Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada,
| |
Collapse
|
20
|
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
|
21
|
Choi SG, Wang Q, Jia J, Chikina M, Pincas H, Dolios G, Sasaki K, Wang R, Minamino N, Salton SRJ, Sealfon SC. Characterization of Gonadotrope Secretoproteome Identifies Neurosecretory Protein VGF-derived Peptide Suppression of Follicle-stimulating Hormone Gene Expression. J Biol Chem 2016; 291:21322-21334. [PMID: 27466366 DOI: 10.1074/jbc.m116.740365] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Indexed: 01/21/2023] Open
Abstract
Reproductive function is controlled by the pulsatile release of hypothalamic gonadotropin-releasing hormone (GnRH), which regulates the expression of the gonadotropins luteinizing hormone and FSH in pituitary gonadotropes. Paradoxically, Fshb gene expression is maximally induced at lower frequency GnRH pulses, which provide a very low average concentration of GnRH stimulation. We studied the role of secreted factors in modulating gonadotropin gene expression. Inhibition of secretion specifically disrupted gonadotropin subunit gene regulation but left early gene induction intact. We characterized the gonadotrope secretoproteome and global mRNA expression at baseline and after Gαs knockdown, which has been found to increase Fshb gene expression (1). We identified 1077 secreted proteins or peptides, 19 of which showed mRNA regulation by GnRH or/and Gαs knockdown. Among several novel secreted factors implicated in Fshb gene regulation, we focused on the neurosecretory protein VGF. Vgf mRNA, whose gene has been implicated in fertility (2), exhibited high induction by GnRH and depended on Gαs In contrast with Fshb induction, Vgf induction occurred preferentially at high GnRH pulse frequency. We hypothesized that a VGF-derived peptide might regulate Fshb gene induction. siRNA knockdown or extracellular immunoneutralization of VGF augmented Fshb mRNA induction by GnRH. GnRH stimulated the secretion of the VGF-derived peptide NERP1. NERP1 caused a concentration-dependent decrease in Fshb gene induction. These findings implicate a VGF-derived peptide in selective regulation of the Fshb gene. Our results support the concept that signaling specificity from the cell membrane GnRH receptor to the nuclear Fshb gene involves integration of intracellular signaling and exosignaling regulatory motifs.
Collapse
Affiliation(s)
| | - Qian Wang
- From the Departments of Neurology and
| | | | | | | | | | - Kazuki Sasaki
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center Research Institute, Osaka 565-8565, Japan
| | | | - Naoto Minamino
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center Research Institute, Osaka 565-8565, Japan
| | - Stephen R J Salton
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029 and
| | - Stuart C Sealfon
- From the Departments of Neurology and Center for Advanced Research on Diagnostic Assays, and
| |
Collapse
|
22
|
Thompson IR, Ciccone NA, Zhou Q, Xu S, Khogeer A, Carroll RS, Kaiser UB. GnRH Pulse Frequency Control of Fshb Gene Expression Is Mediated via ERK1/2 Regulation of ICER. Mol Endocrinol 2016; 30:348-60. [PMID: 26835742 DOI: 10.1210/me.2015-1222] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The pulsatile release of GnRH regulates the synthesis and secretion of pituitary FSH and LH. Two transcription factors, cAMP-response element-binding protein (CREB) and inducible cAMP early repressor (ICER), have been implicated in the regulation of rat Fshb gene expression. We previously showed that the protein kinase A pathway mediates GnRH-stimulated CREB activation. We hypothesized that CREB and ICER are activated by distinct signaling pathways in response to pulsatile GnRH to modulate Fshb gene expression, which is preferentially stimulated at low vs high pulse frequencies. In the LβT2 gonadotrope-derived cell line, GnRH stimulation increased ICER mRNA and protein. Blockade of ERK activation with mitogen-activated protein kinase kinase I/II (MEKI/II) inhibitors significantly attenuated GnRH induction of ICER mRNA and protein, whereas protein kinase C, calcium/calmodulin-dependent protein kinase II, and protein kinase A inhibitors had minimal effects. GnRH also stimulated ICER in primary mouse pituitary cultures, attenuated similarly by a MEKI/II inhibitor. In a perifusion paradigm, MEKI/II inhibition in LβT2 cells stimulated with pulsatile GnRH abrogated ICER induction at high GnRH pulse frequencies, with minimal effect at low frequencies. MEKI/II inhibition reduced GnRH stimulation of Fshb at high and low pulse frequencies, suggesting that the ERK pathway has additional effects on GnRH regulation of Fshb, beyond those mediated by ICER. Indeed, induction of the activating protein 1 proteins, cFos and cJun, positive modulators of Fshb transcription, by pulsatile GnRH was also abrogated by inhibition of the MEK/ERK signaling pathway. Collectively, these studies indicate that the signaling pathways mediating GnRH activation of CREB and ICER are distinct, contributing to the decoding of the pulsatile GnRH to regulate FSHβ expression.
Collapse
Affiliation(s)
- Iain R Thompson
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Nick A Ciccone
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Qiongjie Zhou
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Shuyun Xu
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Ahmad Khogeer
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Rona S Carroll
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
23
|
Candlish M, Angelis RD, Götz V, Boehm U. Gene Targeting in Neuroendocrinology. Compr Physiol 2015; 5:1645-76. [DOI: 10.1002/cphy.c140079] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Fortin J, Ongaro L, Li Y, Tran S, Lamba P, Wang Y, Zhou X, Bernard DJ. Minireview: Activin Signaling in Gonadotropes: What Does the FOX say… to the SMAD? Mol Endocrinol 2015; 29:963-77. [PMID: 25942106 DOI: 10.1210/me.2015-1004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The activins were discovered and named based on their abilities to stimulate FSH secretion and FSHβ (Fshb) subunit expression by pituitary gonadotrope cells. According to subsequent in vitro observations, activins also stimulate the transcription of the GnRH receptor (Gnrhr) and the activin antagonist, follistatin (Fst). Thus, not only do activins stimulate FSH directly, they have the potential to regulate both FSH and LH indirectly by modulating gonadotrope sensitivity to hypothalamic GnRH. Moreover, activins may negatively regulate their own actions by stimulating the production of one of their principal antagonists. Here, we describe our current understanding of the mechanisms through which activins regulate Fshb, Gnrhr, and Fst transcription in vitro. The activin signaling molecules SMAD3 and SMAD4 appear to partner with the winged-helix/forkhead transcription factor, forkhead box L2 (FOXL2), to regulate expression of all 3 genes. However, in vivo data paint a different picture. Although conditional deletion of Foxl2 and/or Smad4 in murine gonadotropes produces impairments in FSH synthesis and secretion as well as in pituitary Fst expression, Gnrhr mRNA levels are either unperturbed or increased in these animals. Surprisingly, gonadotrope-specific deletion of Smad3 alone or with Smad2 does not impair FSH production or fertility; however, mice harboring these mutations may express a DNA binding-deficient, but otherwise functional, SMAD3 protein. Collectively, the available data firmly establish roles for FOXL2 and SMAD4 in Fshb and Fst expression in gonadotrope cells, whereas SMAD3's role requires further investigation. Gnrhr expression, in contrast, appears to be FOXL2, SMAD4, and, perhaps, activin independent in vivo.
Collapse
Affiliation(s)
- Jérôme Fortin
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Luisina Ongaro
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Yining Li
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Stella Tran
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Pankaj Lamba
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Ying Wang
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Xiang Zhou
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| |
Collapse
|
25
|
Boerboom D, Kumar V, Boyer A, Wang Y, Lambrot R, Zhou X, Rico C, Boehm U, Paquet M, Céleste C, Kimmins S, Bernard DJ. β-catenin stabilization in gonadotropes impairs FSH synthesis in male mice in vivo. Endocrinology 2015; 156:323-33. [PMID: 25343272 PMCID: PMC4272392 DOI: 10.1210/en.2014-1296] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although classically considered a WNT signaling intermediary, β-catenin (CTNNB1) can also mediate GnRH induction of gonadotropin β-subunit (Fshb and Lhb) transcription in the murine gonadotrope-like cell line LβT2. Here, we assessed CTNNB1's role in gonadotropin synthesis in vivo. We used a Cre/lox approach to introduce both gain- and loss-of-function mutations in the murine Ctnnb1 gene in gonadotrope cells. Gonadotropin production and fertility were normal in Ctnnb1 knockout mice. Similarly, females harboring a deletion of exon 3 of Ctnnb1, which stabilizes the resulting CTNNB1 protein, showed normal fertility and gonadotropin synthesis. Interestingly, males with the activating CTNNB1-Δexon 3 mutation exhibited 50% reductions in FSH synthesis and secretion, without a corresponding change in LH. This selective regulation of FSH suggested an alteration in the activin/inhibin/follistatin system. Indeed, CTNNB1-Δexon 3 males showed a 60% increase in serum inhibin B levels, and in culture, their pituitaries exhibited a greater sensitivity to exogenous inhibin than controls. At the same time, pituitary, but not testicular, follistatin (Fst) expression was increased significantly in these mice. Castration normalized FSH levels in CTNNB1-Δexon 3 males to those seen in castrated controls. Paradoxically, pituitaries from CTNNB1-Δexon 3 males exhibited greater basal and activin-stimulated FSH synthesis in vitro. Similarly, CTNNB1-Δexon 3 overexpression potentiated activin A-induced murine Fshb promoter activity in LβT2 cells. Together, these results indicate that CTNNB1 is dispensable for gonadotropin synthesis in vivo. However, sustained CTNNB1 signaling potentiates activin-induced Fshb expression in gonadotropes, but this effect is overcome in vivo by enhanced inhibin feedback sensitivity and Fst expression.
Collapse
Affiliation(s)
- Derek Boerboom
- Departments of Pharmacology and Therapeutics (V.K., Y.W., X.Z., S.K., D.J.B.) and Animal Science (R.L., S.K.), McGill University, Montréal, Canada H3G 1Y6; Département de Biomédecine Vétérinaire (D.B., V.K., A.B., C.R., C.C.) and Département de Pathologie et Microbiologie (M.P.), Université de Montréal, St-Hyacinthe, Canada J2S 7C6; and Department of Pharmacology and Toxicology (U.B.), University of Saarland School of Medicine, 66421 Homburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Fortin J, Boehm U, Deng CX, Treier M, Bernard DJ. Follicle-stimulating hormone synthesis and fertility depend on SMAD4 and FOXL2. FASEB J 2014; 28:3396-410. [PMID: 24739304 DOI: 10.1096/fj.14-249532] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Follicle-stimulating hormone (FSH) is an essential regulator of gonadal function and fertility. Loss-of-function mutations in the FSHB/Fshb gene cause hypogonadotropic hypogonadism in humans and mice. Both gonadotropin-releasing hormone (GnRH) and activins, members of the transforming growth factor β (TGFβ) superfamily, stimulate FSH synthesis; yet, their relative roles and mechanisms of action in vivo are unknown. Here, using conditional gene-targeting, we show that the canonical mediator of TGFβ superfamily signaling, SMAD4, is absolutely required for normal FSH synthesis in both male and female mice. Moreover, when the Smad4 gene is ablated in combination with its DNA binding cofactor Foxl2 in gonadotrope cells, mice make essentially no FSH and females are sterile. Indeed, the phenotype of these animals is remarkably similar to that of Fshb-knockout mice. Not only do these results establish SMAD4 and FOXL2 as essential master regulators of Fshb transcription in vivo, they also suggest that activins, or related ligands, could play more important roles in FSH synthesis than GnRH.
Collapse
Affiliation(s)
- Jérôme Fortin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada;
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology, University of Saarland School of Medicine, Homburg, Germany
| | - Chu-Xia Deng
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA; and
| | - Mathias Treier
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada;
| |
Collapse
|