1
|
Cannarella R, Crafa A, Sapienza S, Curto R, Caruso MM, Condorelli RA, La Vignera S, Calogero AE. Testicular function in postpubertal patients with growth hormone deficiency: A prospective controlled study. J Clin Transl Endocrinol 2025; 39:100383. [PMID: 39897110 PMCID: PMC11787440 DOI: 10.1016/j.jcte.2025.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/06/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
Background We have previously shown that treatment with recombinant human growth hormone (GH) influences testicular growth in children with GH deficiency (GHD) and have suggested that GH plays a role in testicular growth in childhood. Little evidence is available on testicular function in post-pubertal GHD patients. Objective This prospective controlled study was undertaken to evaluate testicular function in patients with GHD. Patients and Methods Post-pubertal patients with non-syndromic GHD over the age of 16 years were enrolled. Each patient underwent to the assessment of serum levels of gonadotropins and total testosterone (TT), conventional sperm parameters, and testicular volume (TV) measured by ultrasound examination. Age-matched healthy subjects served as controls. Patients with disorders capable of interfering with testicular function were excluded. Results 26 patients with GHD and 25 age-matched post-pubertal controls were enrolled. They did not differ in serum luteinizing hormone, follicle-stimulating hormone, and TT levels. However, GHD patients had lower semen volume, total sperm count, progressive motility, and total motility values, and a higher prevalence of oligozoospermia compared to controls. No difference was found in sperm concentration and normal morphology. Importantly, GHD patients had lower TV, and a higher prevalence of testicular hypotrophy. Conclusion This is the first evidence of mildly impaired sperm parameters and TV in GHD patients compared to healthy controls. The integrity of the GH-IGF1 axis in prepuberty is important for achieving normal testicular function in adulthood. Evaluating testicular growth over time in GHD children and measuring TV and sperm parameters in postpubertal GHD boys is advisable.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sabrina Sapienza
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Manuela M. Caruso
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A. Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
2
|
Calcaterra V, Tiranini L, Magenes VC, Rossi V, Cucinella L, Nappi RE, Zuccotti G. Impact of Obesity on Pubertal Timing and Male Fertility. J Clin Med 2025; 14:783. [PMID: 39941454 PMCID: PMC11818283 DOI: 10.3390/jcm14030783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Childhood obesity has profound effects on puberty in boys and girls, altering its timing, progression, and associated hormonal changes. Also, later male fertility could be impaired by childhood and pubertal obesity in light of the impact of inflammatory markers on semen quality. The aim of this narrative review is to explore the intricate relationship between childhood obesity and its impact on pubertal development and fertility, with a specific focus on boys. Such a relationship between obesity and pubertal timing in males is highly influenced by metabolic, hormonal, genetic, epigenetic, and environmental factors. While many studies suggest that obesity accelerates pubertal onset in boys, some studies do not confirm these findings, especially in cases of severe obesity. In fact, delayed puberty has also been reported in certain instances. Obesity influences fertility through different central and peripheral processes, including an altered endocrine milieu, inflammatory environment, and epigenetic modifications that alter semen quality and vitality, leading to subfertility or infertility. The early identification and management of potential issues associated with obesity are crucial for ensuring optimal reproductive health in adulthood. Further research is essential to clarify these associations and to develop targeted interventions aimed at preventing the negative health outcomes associated with obesity-related disruptions in puberty and fertility.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (V.R.); (G.Z.)
| | - Lara Tiranini
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (L.T.); (L.C.); (R.E.N.)
| | | | - Virginia Rossi
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (V.R.); (G.Z.)
| | - Laura Cucinella
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (L.T.); (L.C.); (R.E.N.)
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Rossella Elena Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (L.T.); (L.C.); (R.E.N.)
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (V.R.); (G.Z.)
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milano, Italy
| |
Collapse
|
3
|
Khan MZ, Zugaza JL, Torres Aleman I. The signaling landscape of insulin-like growth factor 1. J Biol Chem 2025; 301:108047. [PMID: 39638246 PMCID: PMC11748690 DOI: 10.1016/j.jbc.2024.108047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
The sheer amplitude of biological actions of insulin-like growth factor I (IGF-1) affecting all types of cells in all tissues suggests a vast signaling landscape for this ubiquitous humoral signal. While the canonical signaling pathways primarily involve the Ras/MAPK and PI3K/AKT cascades, the evolutionary conservation of insulin-like peptides (ILPs) and their pathways hints at the potential for novel functions to emerge over time. Indeed, the evolutionary trajectory of ILPs opens the possibility of either novel functions for these two pathways, novel downstream routes, or both. Evidence supporting this notion includes observations of neofunctionalization in bony fishes or crustaceans, and the involvement of ILPs pathways in invertebrate eusociality or in vertebrate bone physiology, respectively. Such evolutionary processes likely contribute to the rich diversity of ILPs signaling observed today. Moreover, the interplay between conserved signaling pathways, such as those implicated in aging (predominantly involving the PI3K-AKT route), and lesser known pathways, such as those mediated by biased G-protein coupled receptors and others even less known, may underpin the context-dependent actions characteristic of ILPs signaling. While canonical IGF-1 signaling is often assumed to account for the intracellular pathways utilized by this growth factor, a comprehensive analysis of all the pathways mediated by the IGF-1 receptor (IGF-1R) remains lacking. This review aims to explore both canonical and non-canonical routes of IGF-1R action across various cell types, offering a detailed examination of the mechanisms underlying IGF-1 signaling and highlighting the significant gaps in our current understanding.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain
| | - Jose Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque Science Foundation, Bilbao, Spain
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain; Ikerbasque Science Foundation, Bilbao, Spain.
| |
Collapse
|
4
|
Guo X, Su L, Shi M, Sun L, Chen W, Geng J, Li J, Zong Y, He Z, Du R. Network Pharmacology and Transcriptomics to Explore the Pharmacological Mechanisms of 20(S)-Protopanaxatriol in the Treatment of Depression. Int J Mol Sci 2024; 25:7574. [PMID: 39062817 PMCID: PMC11276827 DOI: 10.3390/ijms25147574] [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: 04/25/2024] [Revised: 06/15/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Depression is one of the most common psychological disorders nowadays. Studies have shown that 20(S)-protopanaxatriol (PPT) can effectively improve depressive symptoms in mice. However, its mechanism needs to be further explored. In this study, we used an integrated approach combining network pharmacology and transcriptomics to explore the potential mechanisms of PPT for depression. First, the potential targets and pathways of PPT treatment of depression were screened through network pharmacology. Secondly, the BMKCloud platform was used to obtain brain tissue transcription data of chronic unpredictable mild stress (CUMS) model mice and screen PPT-altered differential expression genes (DEGs). Gene ontology (GO) analysis and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed using network pharmacology and transcriptomics. Finally, the above results were verified by molecular docking, Western blotting, and quantitative real-time polymerase chain reaction (qRT-PCR). In this study, we demonstrated that PPT improved depression-like behavior and brain histopathological changes in CUMS mice, downregulated nitric oxide (NO) and interleukin-6 (IL-6) levels, and elevated serum levels of 5-hydroxytryptamine (5-HT) and brain-derived neurotrophic factor (BDNF) after PPT treatment compared to the CUMS group. Eighty-seven potential targets and 350 DEGs were identified by network pharmacology and transcriptomics. Comprehensive analysis showed that transthyretin (TTR), klotho (KL), FOS, and the phosphatidylinositol 3-kinase-protein kinase B (PI3K-AKT) signaling pathway were closely associated with the therapeutic effects of PPT. Molecular docking results showed that PPT had a high affinity for PI3K, AKT, TTR, KL, and FOS targets. Gene and protein level results showed that PPT could increase the expression of PI3K, phosphorylation of PI3K (p-PI3K), AKT, phosphorylation of AKT (p-AKT), TTR, and KL and inhibit the expression level of FOS in the brain tissue of depressed mice. Our data suggest that PPT may achieve the treatment of depression by inhibiting the expression of FOS, enhancing the expression of TTR and KL, and modulating the PI3K-AKT signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhongmei He
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; (X.G.); (L.S.); (M.S.); (L.S.); (W.C.); (J.G.); (J.L.); (Y.Z.)
| | - Rui Du
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; (X.G.); (L.S.); (M.S.); (L.S.); (W.C.); (J.G.); (J.L.); (Y.Z.)
| |
Collapse
|
5
|
Cannarella R, Curto R, Condorelli RA, Lundy SD, La Vignera S, Calogero AE. Molecular insights into Sertoli cell function: how do metabolic disorders in childhood and adolescence affect spermatogonial fate? Nat Commun 2024; 15:5582. [PMID: 38961093 PMCID: PMC11222552 DOI: 10.1038/s41467-024-49765-1] [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: 08/07/2023] [Accepted: 06/12/2024] [Indexed: 07/05/2024] Open
Abstract
Male infertility is a major public health concern globally with unknown etiology in approximately half of cases. The decline in total sperm count over the past four decades and the parallel increase in childhood obesity may suggest an association between these two conditions. Here, we review the molecular mechanisms through which obesity during childhood and adolescence may impair future testicular function. Several mechanisms occurring in obesity can interfere with the delicate metabolic processes taking place at the testicular level during childhood and adolescence, providing the molecular substrate to hypothesize a causal relationship between childhood obesity and the risk of low sperm counts in adulthood.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Scott D Lundy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
6
|
Lou Y, Liang Q, Xin L, Ren M, Hang Q, Qin F, Xiong Z. Integrated untargeted and targeted testicular metabolomics to reveal the regulated mechanism of Gushudan on the hypothalamic-pituitary-gonadal axis of kidney-yang-deficiency-syndrome rats. Biomed Chromatogr 2024; 38:e5872. [PMID: 38638009 DOI: 10.1002/bmc.5872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 04/20/2024]
Abstract
Modern studies have shown that neuroendocrine disorders caused by the dysfunction of the hypothalamic-pituitary-gonadal (HPG) axis are one of the important pathogenetic mechanisms of kidney-yang-deficiency-syndrome (KYDS). The preventive effect of Gushudan on KYDS has been reported, but its regulatory mechanisms on the HPG axis have not been elucidated. In this study, we developed an integrated untargeted and targeted metabolomics analysis strategy to investigate the regulatory mechanism of Gushudan on the HPG axis in rats with KYDS. In untargeted metabolomics, we screened 14 potential biomarkers such as glycine, lysine, and glycerol that were significantly associated with the HPG axis. To explore the effect of changes in the levels of potential biomarkers on KYDS, all of them were quantified in targeted metabolomics. With the quantitative results, correlations between potential biomarkers and testosterone, a functional indicator of the HPG axis, were explored. The results showed that oxidative stress, inflammatory response, and energy depletion, induced by metabolic disorders in rats, were responsible for the decrease in testosterone levels. Gushudan improves metabolic disorders and restores testosterone levels, thus restoring HPG axis dysfunction. This finding elucidates the special metabolic characteristics of KYDS and the therapeutic mechanism of Gushudan from a new perspective.
Collapse
Affiliation(s)
- Yanwei Lou
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, China
| | - Qinghua Liang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, China
| | - Ling Xin
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, China
| | - Mengxin Ren
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, China
| | - Qian Hang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, China
| | - Feng Qin
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, China
| | - Zhili Xiong
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, China
| |
Collapse
|
7
|
Cannarella R, Crafa A, Curto R, Condorelli RA, La Vignera S, Calogero AE. Obesity and male fertility disorders. Mol Aspects Med 2024; 97:101273. [PMID: 38593513 DOI: 10.1016/j.mam.2024.101273] [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: 02/22/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
Often associated with obesity, male infertility represents a widespread condition that challenges the wellbeing of the couple. In this article, we provide a comprehensive and critical analysis of studies exploring the association between obesity and male reproductive function, to evaluate the frequency of this association, and establish the effects of increased body weight on conventional and biofunctional sperm parameters and infertility. In an attempt to find possible molecular markers of infertility in obese male patients, the numerous mechanisms responsible for infertility in overweight/obese patients are reviewed in depth. These include obesity-related functional hypogonadism, insulin resistance, hyperinsulinemia, chronic inflammation, adipokines, irisin, gut hormones, gut microbiome, and sperm transcriptome. According to meta-analytic evidence, excessive body weight negatively influences male reproductive health. This can occurr through a broad array of molecular mechanisms. Some of these are not yet fully understood and need to be further elucidated in the future. A better understanding of the effects of metabolic disorders on spermatogenesis and sperm fertilizing capacity is very useful for identifying new diagnostic markers and designing therapeutic strategies for better clinical management of male infertility.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
8
|
Ye H, Soede NM, Kemp B, Wang J, Fleuren M, Laurenssen B, Bouwman E, Langendijk P. Lactation body condition loss impaired conceptus development and plasma progesterone concentration at day 8 post-ovulation in primiparous sows. Theriogenology 2024; 218:174-182. [PMID: 38330861 DOI: 10.1016/j.theriogenology.2024.02.003] [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: 12/30/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 02/10/2024]
Abstract
The current study investigated effects of dietary amino acid (AA) availability on lactational body condition loss and metabolic status, in relation to reproductive parameters after weaning up to Day 8 post-ovulation. Primiparous sows (n = 35) were allocated to one of two lactation diets containing either low crude protein (CP, 140 g/kg) with a low percentage (8%) of slow protein in total protein (LL, n = 18) or high CP (180 g/kg) with a high (16%) percentage of slow protein (HH, n = 17). The HH diet was expected to improve AA utilization by supplying more AA, in a more gradual fashion. The diets did not affect sow body condition loss during lactation, while the HH diet tended to increase litter weight gain during the week 3 of lactation (Δ = 1.3 kg, P = 0.09). On Day 14 post-farrowing, HH diet led to higher plasma urea both pre-feeding and post-feeding (Δ = 2.3 mmol/L, P < 0.01, Δ = 2.4 mmol/L, P < 0.01, respectively), whilst plasma creatinine, NEFA and IGF-1 were similar. No dietary effects on reproductive parameters were found, however several relationships were found between body condition and reproductive parameters. Sows with higher body weight on Day 1 or Day 21 post-farrowing had greater follicle size on Day 3 post-weaning (β = 0.03 mm/kg, P < 0.01, β = 0.04 mm/kg, P < 0.01, respectively). At Day 8 post-ovulation, plasma progesterone concentration was negatively related to loin muscle loss (β = -0.67 ng/ml · mm-1, P = 0.02), backfat loss (β = -2.33 ng/ml · mm-1, P = 0.02), and estimated body fat loss (β = -0.67 ng/ml · mm-1, P = 0.02). Both plasma progesterone and the number of corpora lutea were positively related to the energy balance during lactation (β = 0.03 ng/ml · ME MJ-1, P = 0.01, β = 0.01 CL/ME MJ, P = 0.02, respectively). The conceptus size at Day 8 post-ovulation was negatively related to body weight loss (β = -0.01 mm/kg, P = 0.01), estimated body fat loss (β = -0.02 mm/kg, P = 0.03) and estimated body protein loss (β = -0.06 mm/kg, P = 0.04), and was positively related to the energy balance during lactation (β = 5.2*10-4 mm/ME MJ, P = 0.01). In conclusion, body protein and fat losses during lactation reduced subsequent plasma progesterone concentration and conceptus development at Day 8 post-ovulation.
Collapse
Affiliation(s)
- Hao Ye
- Adaptation Physiology, Wageningen University and Research, Wageningen, P.O. Box 338, 6700, AH, the Netherlands; State key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 00193, China
| | - Nicoline M Soede
- Adaptation Physiology, Wageningen University and Research, Wageningen, P.O. Box 338, 6700, AH, the Netherlands.
| | - Bas Kemp
- Adaptation Physiology, Wageningen University and Research, Wageningen, P.O. Box 338, 6700, AH, the Netherlands
| | - Junjun Wang
- State key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 00193, China
| | - Marleen Fleuren
- Trouw Nutrition R&D, Stationsstraat, 773811, MH, Amersfoort, the Netherlands
| | - Bjorge Laurenssen
- Adaptation Physiology, Wageningen University and Research, Wageningen, P.O. Box 338, 6700, AH, the Netherlands
| | - Emmy Bouwman
- Trouw Nutrition R&D, Stationsstraat, 773811, MH, Amersfoort, the Netherlands
| | - Pieter Langendijk
- Adaptation Physiology, Wageningen University and Research, Wageningen, P.O. Box 338, 6700, AH, the Netherlands; Trouw Nutrition R&D, Stationsstraat, 773811, MH, Amersfoort, the Netherlands
| |
Collapse
|
9
|
Jiang Y, Gao X, Liu Y, Yan X, Shi H, Zhao R, Chen ZJ, Gao F, Zhao H, Zhao S. Cellular atlases of ovarian microenvironment alterations by diet and genetically-induced obesity. SCIENCE CHINA. LIFE SCIENCES 2024; 67:51-66. [PMID: 37721638 DOI: 10.1007/s11427-023-2360-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/09/2023] [Indexed: 09/19/2023]
Abstract
Obesity, which can arise from genetic or environmental factors, has been shown to cause serious damages to the reproductive system. The ovary, as one of the primary regulators of female fertility, is a complex organ comprised of heterogeneous cell types that work together to maintain a normal ovarian microenvironment (OME). Despite its importance, the effect of obesity on the entire ovary remains poorly documented. In this study, we performed ovary single-cell and nanoscale spatial RNA sequencing to investigate how the OME changed under different kinds of obesity, including high-fat diet (HFD) induced obesity and Leptin ablation induced obesity (OB). Our results demonstrate that OB, but not HFD, dramatically altered the proportion of ovarian granulosa cells, theca-interstitial cells, luteal cells, and endothelial cells. Furthermore, based on the spatial dynamics of follicular development, we defined four subpopulations of granulosa cell and found that obesity drastically disrupted the differentiation of mural granulosa cells from small to large antral follicles. Functionally, HFD enhanced follicle-stimulating hormone (FSH) sensitivity and hormone conversion, while OB caused decreased sensitivity, inadequate steroid hormone conversion, and impaired follicular development. These differences can be explained by the differential expression pattern of the transcription factor Foxo1. Overall, our study provides a powerful and high-resolution resource for profiling obesity-induced OME and offers insights into the diverse effects of obesity on female reproductive disorders.
Collapse
Affiliation(s)
- Yonghui Jiang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, China
| | - Xueying Gao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- Center for reproductive medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127, China
| | - Yue Liu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Xueqi Yan
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Huangcong Shi
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Rusong Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, 250012, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China.
- Center for reproductive medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127, China.
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing, 100101, China.
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China.
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China.
| | - Shigang Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China.
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China.
| |
Collapse
|
10
|
Francoeur L, Scoville DM, Johnson PA. Effect of IGF1 and FSH on the function of granulosa cells from prehierarchal follicles in chickens†. Biol Reprod 2023; 109:498-506. [PMID: 37504508 DOI: 10.1093/biolre/ioad082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/18/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023] Open
Abstract
Insulin-like growth factor 1 (IGF1) is an essential regulator of mammalian follicle development and synergizes with follicle-stimulating hormone (FSH) to amplify its effects. In avian preovulatory follicles, IGF1 increases the expression of genes involved in steroidogenesis and progesterone and inhibin A production. The role of IGF1 in prehierarchal follicles has not been well studied in chickens. The aim of this study was to investigate the role of IGF1 in granulosa cells from prehierarchal follicles and to determine whether IGF1 and FSH synergize to promote follicle development. Granulosa cells of 3-5 and 6-8 mm prehierarchal follicles were cultured with IGF1 (0, 10, 100 ng/mL) in the presence or absence of FSH (0, 10 ng/mL). Cell proliferation, expression of genes important in follicle development (FSHR, IGF1R, AMH, STAR, CYP11A1, INHA, and INHBA), and progesterone production were evaluated following treatment. IGF1 treatment alone significantly increased STAR, CYP11A1, and INHBA mRNA expression and cell proliferation in granulosa cells of 6-8 mm follicles. IGF1 and FSH synergized to increase STAR mRNA expression in 6-8 mm follicles. IGF1 and FSH co-treatment were necessary to increase INHA mRNA expression in 6-8 mm follicles. Although IGF1 significantly increased the expression of genes involved in steroidogenesis, progesterone production in granulosa cells of 6-8 mm follicles was not affected. IGF1 did not affect AMH mRNA expression, although FSH significantly decreased AMH expression in granulosa cells of 3-5 mm follicles. These results suggest that IGF1 may act with FSH to promote follicle selection at the prehierarchal follicle stage.
Collapse
Affiliation(s)
- Laurie Francoeur
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Deena M Scoville
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Patricia A Johnson
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| |
Collapse
|
11
|
Wang Y, Pattarawat P, Zhang J, Kim E, Zhang D, Fang M, Jannaman EA, Yuan Y, Chatterjee S, Kim JYJ, Scott GI, Zhang Q, Xiao S. Effects of Cyanobacterial Harmful Algal Bloom Toxin Microcystin-LR on Gonadotropin-Dependent Ovarian Follicle Maturation and Ovulation in Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:67010. [PMID: 37342990 PMCID: PMC10284350 DOI: 10.1289/ehp12034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/28/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Cyanobacterial harmful algal blooms (CyanoHABs) originate from the excessive growth or bloom of cyanobacteria often referred to as blue-green algae. They have been on the rise globally in both marine and freshwaters in recently years with increasing frequency and severity owing to the rising temperature associated with climate change and increasing anthropogenic eutrophication from agricultural runoff and urbanization. Humans are at a great risk of exposure to toxins released from CyanoHABs through drinking water, food, and recreational activities, making CyanoHAB toxins a new class of contaminants of emerging concern. OBJECTIVES We investigated the toxic effects and mechanisms of microcystin-LR (MC-LR), the most prevalent CyanoHAB toxin, on the ovary and associated reproductive functions. METHODS Mouse models with either chronic daily oral or acute intraperitoneal exposure, an engineered three-dimensional ovarian follicle culture system, and human primary ovarian granulosa cells were tested with MC-LR of various dose levels. Single-follicle RNA sequencing, reverse transcription-quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, western blotting, immunohistochemistry (IHC), and benchmark dose modeling were used to examine the effects of MC-LR on follicle maturation, hormone secretion, ovulation, and luteinization. RESULTS Mice exposed long term to low-dose MC-LR did not exhibit any differences in the kinetics of folliculogenesis, but they had significantly fewer corpora lutea compared with control mice. Superovulation models further showed that mice exposed to MC-LR during the follicle maturation window had significantly fewer ovulated oocytes. IHC results revealed ovarian distribution of MC-LR, and mice exposed to MC-LR had significantly lower expression of key follicle maturation mediators. Mechanistically, in both murine and human granulosa cells exposed to MC-LR, there was reduced protein phosphatase 1 (PP1) activity, disrupted PP1-mediated PI3K/AKT/FOXO1 signaling, and less expression of follicle maturation-related genes. DISCUSSION Using both in vivo and in vitro murine and human model systems, we provide data suggesting that environmentally relevant exposure to the CyanoHAB toxin MC-LR interfered with gonadotropin-dependent follicle maturation and ovulation. We conclude that MC-LR may pose a nonnegligible risk to women's reproductive health by heightening the probability of irregular menstrual cycles and infertility related to ovulatory disorders. https://doi.org/10.1289/EHP12034.
Collapse
Affiliation(s)
- Yingzheng Wang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
- National Institute of Environmental Health Sciences Center for Oceans and Human Health and Climate Change Interactions at the University of South Carolina, Columbia, South Carolina, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, New Jersey, USA
| | - Pawat Pattarawat
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Jiyang Zhang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Eunchong Kim
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Delong Zhang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Mingzhu Fang
- New Jersey Department of Environmental Protection, Trenton, New Jersey, USA
| | | | - Ye Yuan
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado, USA
| | - Saurabh Chatterjee
- Department of Environmental and Occupational Health, University of California, Irvine, Irvine, California, USA
- Division of Infectious Disease, Department of Medicine, University of California, Irvine, Irvine, California, USA
| | - Ji-Yong Julie Kim
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Geoffrey I. Scott
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
- National Institute of Environmental Health Sciences Center for Oceans and Human Health and Climate Change Interactions at the University of South Carolina, Columbia, South Carolina, USA
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
- National Institute of Environmental Health Sciences Center for Oceans and Human Health and Climate Change Interactions at the University of South Carolina, Columbia, South Carolina, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
12
|
Cannarella R, Caruso M, Condorelli RA, Timpanaro TA, Caruso MA, La Vignera S, Calogero AE. Testicular volume in 268 children and adolescents followed-up for childhood obesity-a retrospective cross-sectional study. Eur J Endocrinol 2023; 188:331-342. [PMID: 37127298 DOI: 10.1093/ejendo/lvad033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/21/2022] [Accepted: 03/06/2023] [Indexed: 05/03/2023]
Abstract
CONTEXT Prevalence of obesity in childhood has increased over the past few decades. The impact of obesity and of obesity-related metabolic disorders on testicular growth is unknown. OBJECTIVE To evaluate the impact of obesity, hyperinsulinemia, and insulin resistance on testicular volume (TV) in pre-pubertal (<9 years), peri-pubertal (9-14 years), and post-pubertal (14-16 years) periods. METHODS We collected data on TV, age, standard deviation score (SDS) of the body mass index (BMI), insulin, and fasting glycemia in 268 children and adolescents followed-up for weight control. RESULTS Peri-pubertal boys with normal weight had a significantly higher TV compared to those with overweight or obesity. No difference was found in the other age ranges when data were grouped according to BMI. Pre- and post-pubertal children/adolescents with normal insulin levels had significantly higher TV compared to those with hyperinsulinemia. Peri-pubertal boys with hyperinsulinemia had significantly higher TV compared to those with normal insulin levels. Post-pubertal adolescents with insulin resistance had lower TV and peri-pubertal boys had higher TV compared to those without insulin resistance. No difference was found in pre-puberty. CONCLUSIONS Closer control of the body weight and the associated metabolic alterations in childhood and adolescence may maintain testicular function later in life.
Collapse
Affiliation(s)
- R Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - M Caruso
- Pediatric Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - R A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - T A Timpanaro
- Pediatric Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - M A Caruso
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - S La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - A E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| |
Collapse
|
13
|
Duan L, Calhoun S, Perez RE, Macias V, Mir F, Pergande MR, Gattuso P, Borgia JA, Maki CG. Prolyl Carboxypeptidase Maintains Receptor Tyrosine Kinase Signaling and Is a Potential Therapeutic Target in Triple Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14030739. [PMID: 35159006 PMCID: PMC8833515 DOI: 10.3390/cancers14030739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Triple negative breast cancer (TNBC) is an aggressive cancer type with limited treatment options and poor prognosis. Our research has revealed that a protein called prolylcarboxypeptidase (PRCP) is a potential therapy target for TNBC. We found that high levels of PRCP in tumors coincides with worse prognosis in TNBC patients. Inhibition of PRCP with a small molecule inhibitor blocked TNBC cell and tumor growth and inhibited the activity of several receptor tyrosine kinases (RTKs), proteins that are located on the surface of cells and that are important for cancer development and progression. Our findings suggest that PRCP is a novel prognostic factor for TNBC and that specific inhibitors of PRCP could be developed for TNBC treatment. Abstract TNBC is an aggressive cancer sub-type with limited treatment options and poor prognosis. New therapeutic targets are needed to improve outcomes in TNBC patients. PRCP is a lysosomal serine protease that cleaves peptide substrates when the penultimate amino acid is proline. A role for PRCP in TNBC or other cancers, and its potential as a therapy target has not yet been tested. In the current study, we found high tumor expression of PRCP associates with worse outcome and earlier recurrence in TNBC patients. Knockdown of PRCP or treatment with a small molecule PRCP inhibitor blocked proliferation and survival in TNBC cell lines and inhibited growth of TNBC tumors in mice. Mechanistically, we found PRCP maintains signaling from multiple receptor tyrosine kinases (RTKs), potentially by promoting crosstalk between RTKs and G-protein coupled receptors (GPCRs). Lastly, we found that the PRCP inhibitor caused synergistic killing of TNBC cells when combined with the EGFR and ErbB2 inhibitor lapatinib. Our results suggest that PRCP is potential prognostic marker for TNBC patient outcome and a novel therapeutic target for TNBC treatment.
Collapse
Affiliation(s)
- Lei Duan
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac 507, Chicago, IL 60612, USA; (L.D.); (S.C.); (R.E.P.); (M.R.P.); (J.A.B.)
| | - Sarah Calhoun
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac 507, Chicago, IL 60612, USA; (L.D.); (S.C.); (R.E.P.); (M.R.P.); (J.A.B.)
| | - Ricardo E. Perez
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac 507, Chicago, IL 60612, USA; (L.D.); (S.C.); (R.E.P.); (M.R.P.); (J.A.B.)
| | - Virgilia Macias
- Department of Pathology, University of Illinois at Chicago, 909 S. Wolcott St, Rm 6128, Chicago, IL 60612, USA;
| | - Fatima Mir
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA; (F.M.); (P.G.)
| | - Melissa R. Pergande
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac 507, Chicago, IL 60612, USA; (L.D.); (S.C.); (R.E.P.); (M.R.P.); (J.A.B.)
| | - Paolo Gattuso
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA; (F.M.); (P.G.)
| | - Jeffrey A. Borgia
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac 507, Chicago, IL 60612, USA; (L.D.); (S.C.); (R.E.P.); (M.R.P.); (J.A.B.)
| | - Carl G. Maki
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac 507, Chicago, IL 60612, USA; (L.D.); (S.C.); (R.E.P.); (M.R.P.); (J.A.B.)
- Correspondence: ; Tel.: +312-563-3380
| |
Collapse
|
14
|
Gao L, Gao H, Wang W. Androgens improve ovarian follicle function impaired by glucocorticoids through an androgen-IGF1-FSH synergistic effect. Front Endocrinol (Lausanne) 2022; 13:951928. [PMID: 36339442 PMCID: PMC9627217 DOI: 10.3389/fendo.2022.951928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/29/2022] [Indexed: 12/02/2022] Open
Abstract
High concentrations of glucocorticoids caused by chronic stress are known to affect ovarian function and cause diminished ovarian reserve. Androgens are essential for early-stage ovarian follicle development, but the effects and mechanisms of androgens on follicle development under chronic stress remain unclear. In this study, we aim to investigate the effects of high concentrations of glucocorticoids on the function of in vitro cultured ovarian cells and mouse early-stage ovarian follicles and to validate the hypothesis that androgen-insulin-like growth factor 1 (IGF1)-follicle-stimulating hormone (FSH) synergistic signaling helps to ameliorate the damage caused by high concentrations of glucocorticoids. KGN cells (human granulosa cell line) and mouse primary cells were treated with different concentrations of glucocorticoids, and the cell proliferation, apoptosis, and sex hormone secretion were detected. The effects of glucocorticoid and androgens on IGF1 receptor (IGF1R) and FSH receptor (FSHR) expression in KGN cells were detected by Western blot. Steroidogenic synthase expressions under androgens and androgen-IGF1-FSH combination treatment were examined by qPCR after manipulation using low and high concentrations of glucocorticoids. The mechanism of androgen regulation of IGF1R and FSHR was explored by small interfering RNA (siRNA) and chromatin immunoprecipitation (ChIP)-qPCR. Damage of glucocorticoids and the treatment effects of androgens were further validated in mouse ovarian follicles cultured in vitro. The results demonstrated that prolonged treatment with high-dose glucocorticoids reduced cell viability of granulosa cells, inhibited their sex hormone secretion, and impaired their sensitivity to IGF1 and FSH signaling by affecting IGF1R and FSHR functions. Androgens at an appropriate dose range improved early-stage follicle development and their hormone secretion under high-dose glucocorticoid treatment, which was related to increased transcription of Igf1r and Fshr. This work showed that excessive glucocorticoids impaired ovarian function and validated that balanced concentrations of androgens synergized with IGF1 and FSH to improve the function of early-stage ovarian follicles under conditions of chronic stress.
Collapse
Affiliation(s)
- Lingyun Gao
- Department of Integrated Traditional & Western Medicine, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Integrated Traditional & Western Medicine, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Hongna Gao
- Department of Integrated Traditional & Western Medicine, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Integrated Traditional & Western Medicine, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Wenjun Wang
- Department of Integrated Traditional & Western Medicine, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Integrated Traditional & Western Medicine, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- *Correspondence: Wenjun Wang,
| |
Collapse
|
15
|
Lundin K, Sepponen K, Väyrynen P, Liu X, Yohannes DA, Survila M, Ghimire B, Känsäkoski J, Katayama S, Partanen J, Vuoristo S, Paloviita P, Rahman N, Raivio T, Luiro K, Huhtaniemi I, Varjosalo M, Tuuri T, Tapanainen JS. OUP accepted manuscript. Mol Hum Reprod 2022; 28:6574364. [PMID: 35471239 PMCID: PMC9308958 DOI: 10.1093/molehr/gaac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/11/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- K Lundin
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - K Sepponen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - P Väyrynen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - X Liu
- Molecular Systems Biology Research Group, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
- Proteomics Unit, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
| | - D A Yohannes
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Translational Immunology & Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - M Survila
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - B Ghimire
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - J Känsäkoski
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | - S Katayama
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J Partanen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - S Vuoristo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - P Paloviita
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - N Rahman
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - T Raivio
- Department of Physiology, University of Helsinki, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- New Children's Hospital, Pediatric Research Center, Helsinki University Hospital, HUH, Helsinki, Finland
| | - K Luiro
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - I Huhtaniemi
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Metabolism, Endocrinology and Reproduction, Faculty of Medicine, Hammersmith Campus, Imperial College London, London, UK
| | - M Varjosalo
- Molecular Systems Biology Research Group, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
- Proteomics Unit, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
| | - T Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - J S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, University Hospital of Oulu, University of Oulu, Medical Research Center Oulu and PEDEGO Research Unit, Oulu, Finland
- Corresponding author. Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, PO Box 140, 00029 Helsinki, Finland. Tel: +358-94711; E-mail:
| |
Collapse
|
16
|
Cannarella R, Mancuso F, Arato I, Lilli C, Bellucci C, Gargaro M, Curto R, Aglietti MC, La Vignera S, Condorelli RA, Luca G, Calogero AE. Sperm-carried IGF2 downregulated the expression of mitogens produced by Sertoli cells: A paracrine mechanism for regulating spermatogenesis? Front Endocrinol (Lausanne) 2022; 13:1010796. [PMID: 36523595 PMCID: PMC9744929 DOI: 10.3389/fendo.2022.1010796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Insulin-like growth factor 2 (IGF2) mRNA has been found in human and mouse spermatozoa. It is currently unknown whether the IGF2 protein is expressed in human spermatozoa and, if so, its possible role in the cross-talk between germ and Sertoli cells (SCs) during spermatogenesis. METHODS To accomplish this, we analyzed sperm samples from four consecutive Caucasian men. Furthermore, to understand its role during the spermatogenetic process, porcine SCs were incubated with increasing concentrations (0.33, 3.33, and 10 ng/mL) of recombinant human IGF2 (rhIGF2) for 48 hours. Subsequently, the experiments were repeated by pre-incubating SCs with the non-competitive insulin-like growth factor 1 receptor (IGF1R) inhibitor NVP-AEW541. The following outcomes were evaluated: 1) Gene expression of the glial cell-line derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), and stem cell factor (SCF) mitogens; 2) gene and protein expression of follicle-stimulating hormone receptor (FSHR), anti-Müllerian hormone (AMH), and inhibin B; 3) SC proliferation. RESULTS We found that the IGF2 protein was present in each of the sperm samples. IGF2 appeared as a cytoplasmic protein localized in the equatorial and post-acrosomal segment and with a varying degree of expression in each cell. In SCs, IGF2 significantly downregulated GDNF gene expression in a concentration-dependent manner. FGF2 and SCF were downregulated only by the highest concentration of IGF2. Similarly, IGF2 downregulated the FSHR gene and FSHR, AMH, and inhibin B protein expression. Finally, IGF2 significantly suppressed the SC proliferation rate. All these findings were reversed by pre-incubation with NVP-AEW541, suggesting an effect mediated by the interaction of IGF2 with the IGFR. CONCLUSION In conclusion, sperm IGF2 seems to downregulate the expression of mitogens, which are known to be physiologically released by the SCs to promote gonocyte proliferation and spermatogonial fate adoption. These findings suggest the presence of paracrine regulatory mechanisms acting on the seminiferous epithelium during spermatogenesis, by which germ cells can influence the amount of mitogens released by the SCs, their sensitivity to FSH, and their rate of proliferation.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
- *Correspondence: Rossella Cannarella,
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Maria C. Aglietti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A. Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giovani Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
17
|
Cannarella R, Condorelli RA, Barbagallo F, Aversa A, Calogero AE, La Vignera S. TSH lowering effects of metformin: a possible mechanism of action. J Endocrinol Invest 2021; 44:1547-1550. [PMID: 33058005 PMCID: PMC8195970 DOI: 10.1007/s40618-020-01445-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/07/2020] [Indexed: 01/28/2023]
Abstract
Preliminary clinical evidence suggests that metformin has TSH lowering effects in patients with T2DM and hypothyroidism or in those with TSH serum levels in the upper normal value. Also, metformin may exert a protective role against thyroid nodules growth in patients without insulin-resistance. The cross-talk between tyrosine kinase receptors and the G protein-coupled receptors (which the TSHR belongs to) has been already shown and IRS1 may represent the hub link between TSHR and IR pathways. By influencing IRS1 phosphorylation pattern, metformin may sensitize TSHR to TSH, thus explaining the findings of clinical studies. However, the existence of this molecular pathway must be confirmed through proper studies and further prospective randomized placebo-controlled studies are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- R Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Via S. Sofia 78, 95123, Catania, Italy.
| | - R A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Via S. Sofia 78, 95123, Catania, Italy
| | - F Barbagallo
- Department of Clinical and Experimental Medicine, University of Catania, Via S. Sofia 78, 95123, Catania, Italy
| | - A Aversa
- Department of Experimental and Clinical Medicine, "Magna Graecia" University, Catanzaro, Italy
| | - A E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Via S. Sofia 78, 95123, Catania, Italy
| | - S La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Via S. Sofia 78, 95123, Catania, Italy
| |
Collapse
|
18
|
Costermans NGJ, Teerds KJ, Middelkoop A, Roelen BAJ, Schoevers EJ, van Tol HTA, Laurenssen B, Koopmanschap RE, Zhao Y, Blokland M, van Tricht F, Zak L, Keijer J, Kemp B, Soede NM. Consequences of negative energy balance on follicular development and oocyte quality in primiparous sows†. Biol Reprod 2021; 102:388-398. [PMID: 31504218 PMCID: PMC7016286 DOI: 10.1093/biolre/ioz175] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/22/2019] [Indexed: 01/28/2023] Open
Abstract
Metabolic demands of modern hybrid sows have increased over the years, which increases the chance that sows enter a substantial negative energy balance (NEB) during lactation. This NEB can influence the development of follicles and oocytes that will give rise to the next litter. To study effects of a lactational NEB on follicular development, we used 36 primiparous sows of which 18 were subjected to feed restriction (3.25 kg/day) and 18 were full-fed (6.5 kg/day) during the last 2 weeks of a 24.1 ± 0.3 day lactation. Feed restriction resulted in a 70% larger lactational body weight loss and 76% higher longissimus dorsi depth loss, but similar amounts of backfat loss compared to the full fed sows. These changes were accompanied by lower plasma insulin-like growth factor 1 (IGF1) and higher plasma creatinine levels in the restricted sows from the last week of lactation onward. Ovaries were collected 48 h after weaning. Restricted sows had a lower average size of the 15 largest follicles (−26%) and cumulus–oocyte complexes showed less expansion after 22 h in vitro maturation (−26%). Less zygotes of restricted sows reached the metaphase stage 24 h after in vitro fertilization and showed a higher incidence of polyspermy (+89%). This shows that feed restriction had severe consequences on oocyte developmental competence. Follicular fluid of restricted sows had lower IGF1 (−56%) and steroid levels (e.g., β-estradiol, progestins, and androgens), which indicated that follicles of restricted sows were less competent to produce steroids and growth factors needed for oocytes to obtain full developmental competence.
Collapse
Affiliation(s)
- N G J Costermans
- Human and Animal Physiology, Wageningen University and Research, Wageningen, The Netherlands.,Adaptation Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - K J Teerds
- Human and Animal Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - A Middelkoop
- Adaptation Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - B A J Roelen
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - E J Schoevers
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - H T A van Tol
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - B Laurenssen
- Adaptation Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - R E Koopmanschap
- Adaptation Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Y Zhao
- Human and Animal Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - M Blokland
- Wageningen Food Safety Research (WFSR), Wageningen University & Research, Akkermaalsbos 2, 6708WB Wageningen, The Netherlands
| | - F van Tricht
- Wageningen Food Safety Research (WFSR), Wageningen University & Research, Akkermaalsbos 2, 6708WB Wageningen, The Netherlands
| | - L Zak
- TopigsNorsvin Research Center B. V., Beuningen, The Netherlands
| | - J Keijer
- Human and Animal Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - B Kemp
- Adaptation Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - N M Soede
- Adaptation Physiology, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
19
|
Abstract
Gonadotropins are glycoprotein sex hormones regulating development and reproduction and bind to specific G protein–coupled receptors expressed in the gonads. Their effects on multiple signaling cascades and intracellular events have recently been characterized using novel technological and scientific tools. The impact of allosteric modulators on gonadotropin signaling, the role of sugars linked to the hormone backbone, the detection of endosomal compartments supporting signaling modules, and the dissection of different effects mediated by these molecules are areas that have advanced significantly in the last decade. The classic view providing the exclusive activation of the cAMP/protein kinase A (PKA) and the steroidogenic pathway by these hormones has been expanded with the addition of novel signaling cascades as determined by high-resolution imaging techniques. These new findings provided new potential therapeutic applications. Despite these improvements, unanswered issues of gonadotropin physiology, such as the intrinsic pro-apoptotic potential to these hormones, the existence of receptors assembled as heteromers, and their expression in extragonadal tissues, remain to be studied. Elucidating these issues is a challenge for future research.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
20
|
Pseudo-Starvation Driven Energy Expenditure Negatively Affects Ovarian Follicle Development. Int J Mol Sci 2021; 22:ijms22073557. [PMID: 33808081 PMCID: PMC8036485 DOI: 10.3390/ijms22073557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/23/2021] [Accepted: 03/27/2021] [Indexed: 11/30/2022] Open
Abstract
In the present investigation, we examined whether a change in whole body energy fluxes could affect ovarian follicular development, employing mice ectopically expressing uncoupling protein 1 in skeletal muscle (UCP1-TG). Female UCP1-TG and wild-type (WT) mice were dissected at the age of 12 weeks. Energy intake and expenditure, activity, body weight and length, and body composition were measured. Plasma insulin, glucose, leptin, plasma fibroblast growth factor 21 (FGF21) and plasma insulin-like growth factor 1 (IGF1) levels were analyzed and ovarian follicle and corpus luteum numbers were counted. IGF1 signaling was analyzed by immunohistochemical staining for the activation of insulin receptor substrate 1/2 (IRS1/2) and AKT. UCP1-TG female mice had increased energy expenditure, reduced body size, maintained adiposity, and decreased IGF1 concentrations compared to their WT littermates, while preantral and antral follicle numbers were reduced by 40% and 60%, respectively. Corpora lutea were absent in 40% of the ovaries of UCP1-TG mice. Phospho-IRS1, phospho-AKT -Ser473 and -Thr308 immunostaining was present in the granulosa cells of antral follicles in WT ovaries, but faint to absent in the antral follicles of UCP1-TG mice. In conclusion, the reduction in circulating IGF1 levels due to the ectopic expression of UCP1 is associated with reduced immunostaining of the IRS1-PI3/AKT pathway, which may negatively affect ovarian follicle development and ovulation.
Collapse
|
21
|
Shpakov AO. Improvement Effect of Metformin on Female and Male Reproduction in Endocrine Pathologies and Its Mechanisms. Pharmaceuticals (Basel) 2021; 14:ph14010042. [PMID: 33429918 PMCID: PMC7826885 DOI: 10.3390/ph14010042] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Metformin (MF), a first-line drug to treat type 2 diabetes mellitus (T2DM), alone and in combination with other drugs, restores the ovarian function in women with polycystic ovary syndrome (PCOS) and improves fetal development, pregnancy outcomes and offspring health in gestational diabetes mellitus (GDM) and T2DM. MF treatment is demonstrated to improve the efficiency of in vitro fertilization and is considered a supplementary drug in assisted reproductive technologies. MF administration shows positive effect on steroidogenesis and spermatogenesis in men with metabolic disorders, thus MF treatment indicates prospective use for improvement of male reproductive functions and fertility. MF lacks teratogenic effects and has positive health effect in newborns. The review is focused on use of MF therapy for restoration of female and male reproductive functions and improvement of pregnancy outcomes in metabolic and endocrine disorders. The mechanisms of MF action are discussed, including normalization of metabolic and hormonal status in PCOS, GDM, T2DM and metabolic syndrome and restoration of functional activity and hormonal regulation of the gonadal axis.
Collapse
Affiliation(s)
- Alexander O Shpakov
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 194223 Saint Petersburg, Russia
| |
Collapse
|
22
|
Cannarella R, Musso N, Condorelli RA, Musmeci M, Stefani S, Aversa A, La Vignera S, Calogero AE. The 2039 A/G FSH receptor gene polymorphism influences glucose metabolism in healthy men. Endocrine 2020; 70:629-634. [PMID: 32681384 PMCID: PMC7674314 DOI: 10.1007/s12020-020-02420-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/06/2020] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To assess the role of c. 2039 A/G (p. Asp680Ser) (rs6166) and c. -29 G/A (rs1394205) follicle-stimulating hormone receptor (FSHR) gene single nucleotide polymorphisms (SNPs) in a cohort of healthy men. METHODS One-hundred twenty-seven healthy men underwent evaluation of the anthropometric parameters, assessment of metabolic and lipid profile, measurement FSH serum levels, and genotyping of both the aforementioned FSHR SNPs. Data grouped according to the FSHR rs6166 or rs1394205 genotypes underwent to statistical analysis. MAIN RESULTS The three groups of men for each FSHR SNP did not differ statistically significantly for body mass index and serum FSH levels. As for FSHR rs6166 SNP, glucose levels were significantly lower in men with the GG genotype compared with those with the AA genotype. Men with AG had lower insulin levels and HOMA index values compared with those carrying the genotype AA (p < 0.05). The GG group showed a negative correlation between serum FSH levels and insulin and between serum FSH levels and HOMA index (p < 0.05). In contrast, men grouped according to the FSHR rs1394205 genotype showed no significant difference in blood glucose, serum insulin levels, and HOMA index. The AG group showed a negative correlation between FSH insulin and between serum FSH levels and HOMA index (p < 0.05). CONCLUSIONS Men with the genotype GG of the FSHR rs6166 SNP have lower blood glucose levels than those with the AA genotype. Their FSH levels inversely correlated with insulin and HOMA index. In contrast, the genotype FSHR rs6166 A/G did not reveal any role of FSH on glucose metabolism in healthy men. The inverse relationship between FSH and insulin or HOMA index in the group with the genotype GG of the FSHR rs6166 SNP suggests a possible cross-talk between FSH and insulin.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Nicolò Musso
- Bio-nanotech Research and Innovation Tower (BRIT), University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Marco Musmeci
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Stefania Stefani
- Bio-nanotech Research and Innovation Tower (BRIT), University of Catania, Catania, Italy
| | - Antonio Aversa
- Department of Experimental and Clinical Medicine, "Magna Graecia" University, Catanzaro, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
23
|
Tremblay PG, Sirard MA. Gene analysis of major signaling pathways regulated by gonadotropins in human ovarian granulosa tumor cells (KGN)†. Biol Reprod 2020; 103:583-598. [PMID: 32427331 DOI: 10.1093/biolre/ioaa079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/17/2020] [Accepted: 05/18/2020] [Indexed: 11/13/2022] Open
Abstract
The female reproductive function largely depends on timing and coordination between follicle-stimulating hormone (FSH) and luteinizing hormone. Even though it was suggested that these hormones act on granulosa cells via shared signaling pathways, mainly protein kinases A, B, and C (PKA, PKB, and PKC), there is still very little information available on how these signaling pathways are regulated by each hormone to provide such differences in gene expression throughout folliculogenesis. To obtain a global picture of the principal upstream factors involved in PKA, PKB, and PKC signaling in granulosa cells, human granulosa-like tumor cells (KGN) were treated with FSH or specific activators (forskolin, SC79, and phorbol 12-myristate 13-acetate) for each pathway to analyze gene expression with RNA-seq technology. Normalization and cutoffs (FC 1.5, P ≤ 0.05) revealed 3864 differentially expressed genes between treatments. Analysis of major upstream regulators showed that PKA is a master kinase of early cell differentiation as its activation resulted in the gene expression profile that accompanies granulosa cell differentiation. Our data also revealed that the activation of PKC in granulosa cells is also a strong differentiation signal that could control "advanced" differentiation in granulosa cells and the inflammatory cascade that occurs in the dominant follicle. According to our results, PKB activation provides support for PKA-stimulated gene expression and is also involved in granulosa cell survival throughout follicular development. Taken together, our results provide new information on PKA, PKB, and PKC signaling pathways and their roles in stimulating a follicle at the crossroad between maturation/ovulation and atresia.
Collapse
Affiliation(s)
- Patricia G Tremblay
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des sciences de l'agriculture et de l'alimentation, Département des Sciences animales, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Marc-André Sirard
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des sciences de l'agriculture et de l'alimentation, Département des Sciences animales, Université Laval, Québec, QC, G1V 0A6, Canada
| |
Collapse
|
24
|
Plewes MR, Hou X, Talbott HA, Zhang P, Wood JR, Cupp AS, Davis JS. Luteinizing hormone regulates the phosphorylation and localization of the mitochondrial effector dynamin-related protein-1 (DRP1) and steroidogenesis in the bovine corpus luteum. FASEB J 2020; 34:5299-5316. [PMID: 32077149 DOI: 10.1096/fj.201902958r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/31/2022]
Abstract
The corpus luteum is an endocrine gland that synthesizes and secretes progesterone. Luteinizing hormone (LH) activates protein kinase A (PKA) signaling in luteal cells, increasing delivery of substrate to mitochondria for progesterone production. Mitochondria maintain a highly regulated equilibrium between fusion and fission in order to sustain biological function. Dynamin-related protein 1 (DRP1), is a key mediator of mitochondrial fission. The mechanism by which DRP1 is regulated in the ovary is largely unknown. We hypothesize that LH via PKA differentially regulates the phosphorylation of DRP1 on Ser616 and Ser637 in bovine luteal cells. In primary cultures of steroidogenic small luteal cells (SLCs), LH, and forskolin stimulated phosphorylation of DRP1 (Ser 637), and inhibited phosphorylation of DRP1 (Ser 616). Overexpression of a PKA inhibitor blocked the effects of LH and forskolin on DRP1 phosphorylation. In addition, LH decreased the association of DRP1 with the mitochondria. Genetic knockdown of the DRP1 mitochondria receptor, and a small molecule inhibitor of DRP1 increased basal and LH-induced progesterone production. Studies with a general Dynamin inhibitor and siRNA knockdown of DRP1 showed that DRP1 is required for optimal LH-induced progesterone biosynthesis. Taken together, the findings place DRP1 as an important target downstream of PKA in steroidogenic luteal cells.
Collapse
Affiliation(s)
- Michele R Plewes
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA.,Nebraska Western Iowa Veterans Health Care System, Omaha, NE, USA
| | - Xiaoying Hou
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA
| | - Heather A Talbott
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA
| | - Pan Zhang
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA
| | - Jennifer R Wood
- Department of Animal Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Andrea S Cupp
- Department of Animal Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA.,Nebraska Western Iowa Veterans Health Care System, Omaha, NE, USA
| |
Collapse
|
25
|
Chu YL, Xu YR, Yang WX, Sun Y. The role of FSH and TGF-β superfamily in follicle atresia. Aging (Albany NY) 2019; 10:305-321. [PMID: 29500332 PMCID: PMC5892684 DOI: 10.18632/aging.101391] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/23/2018] [Indexed: 01/02/2023]
Abstract
Most of the mammalian follicles undergo a degenerative process called “follicle atresia”. Apoptosis of granulosa cells is the main characteristic of follicle atresia. Follicle stimulating hormone (FSH) and the transforming growth factor β (TGF-β) superfamily have important regulatory functions in this process. FSH activates protein kinase A and cooperating with insulin receptor substrates, it promotes the PI3K/Akt pathway which weakens apoptosis. Both Smad or non-Smad signaling of the transforming growth factor β superfamily seem to be related to follicle atresia, and the effect of several important family members on follicle atresia is concluded in this article. FSH and TGF-β are likely to mutually influence each other and what we have already known about the possible underlying molecular mechanism is also discussed below.
Collapse
Affiliation(s)
- Yu-Lan Chu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ya-Ru Xu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Sun
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
26
|
Cannarella R, Arato I, Condorelli RA, Mongioì LM, Lilli C, Bellucci C, La Vignera S, Luca G, Mancuso F, Calogero AE. Effects of Insulin on Porcine Neonatal Sertoli Cell Responsiveness to FSH In Vitro. J Clin Med 2019; 8:jcm8060809. [PMID: 31174276 PMCID: PMC6617126 DOI: 10.3390/jcm8060809] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022] Open
Abstract
There is ongoing debate as to whether the decline of sperm production in recent times may be related to a parallel increase in the rate of obesity and diabetes. Lower anti-Müllerian hormone (AMH) and inhibin B secretion have been observed in young hyperinsulinemic patients compared to healthy controls, suggesting a Sertoli cell (SC) dysfunction. The pathophysiological mechanisms underlying SC dysfunction in these patients are poorly understood. To the best of our knowledge, no evidence is available on the effects of insulin on SC function. Therefore, this study was undertaken to assess the effects of insulin on basal and follicle-stimulating hormone (FSH)-stimulated SC function in vitro. To accomplish this, we evaluated the expression of AMH, inhibin B and FSHR genes, the secretion of AMH and inhibin B and the phosphorylation of AKT473 and SC proliferation on neonatal porcine SC after incubation with FSH and/or insulin. We found that similar to FSH, the expression and secretion of AMH is suppressed by insulin. Co-incubation with FSH and insulin decreased AMH secretion significantly more than with FSH alone. Insulin had no effect on the expression and secretion of the inhibin B gene, but co-incubation with FSH and insulin had a lower effect on inhibin B secretion than that found with FSH alone. FSH and/or insulin increased AKT473 phosphorylation and SC proliferation. In conclusion, the results of this study showed that insulin modulates SC function. We hypothesize that hyperinsulinemia may therefore influence testicular function even before puberty begins. Therefore, particular care should be taken to avoid the onset of hyperinsulinemia in children to prevent a future deleterious effect on fertility.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Iva Arato
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Laura M Mongioì
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Cinzia Lilli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Catia Bellucci
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Francesca Mancuso
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
27
|
Cannarella R, Arato I, Condorelli RA, Luca G, Barbagallo F, Alamo A, Bellucci C, Lilli C, La Vignera S, Calafiore R, Mancuso F, Calogero AE. The IGF1 Receptor Is Involved in Follicle-Stimulating Hormone Signaling in Porcine Neonatal Sertoli Cells. J Clin Med 2019; 8:jcm8050577. [PMID: 31035547 PMCID: PMC6571966 DOI: 10.3390/jcm8050577] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
Experimental evidence has shown that the IGF1 receptor (IGF1R) is involved in testicular development during embryogenesis. More recently, data gathered from mice granulosa cells and zebrafish spermatogonia suggest that IGF1R has a role in Follicle-stimulating hormone (FSH) signaling. No evidence has been reported on this matter in Sertoli cells (SCs) so far. The aim of the study was to evaluate the role, if any, of the IGF1R in FSH signaling in SCs. The effects of FSH exposure on myosin-phosphatase 1 (MYPT1), ERK 1/2, AKT308, AKT473, c-Jun N-terminal kinase (JNK) phosphorylation and on anti-Müllerian hormone (AMH), inhibin B and FSH receptor (FSHR) mRNA levels were assessed with and without the IGF1R inhibitor NVP-AEW541 in purified and functional porcine neonatal SCs. Pre-treatment with NVP-AEW541 inhibited the FSH-induced MYPT1 and ERK 1/2 phosphorylation, decreased the FSH-dependent Protein kinase B (AKT)308 phosphorylation, but did not affect the FSH-induced AKT473 and JNK phosphorylation rate. It also interfered with the FSH-induced AMH and FSHR down-regulation. No influence was observed on the FSH-stimulated Inhibin B gene expression. Conclusion. These findings support the role of theIGF1R in FSH signaling in porcine SCs. The possible influence of IGF1 stimulation on the FSH-mediated effects on SCs should be further explored.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Iva Arato
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Federica Barbagallo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Angela Alamo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Catia Bellucci
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Cinzia Lilli
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | | | - Francesca Mancuso
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
28
|
Cannarella R, Condorelli RA, La Vignera S, Calogero AE. Commentary: Molecular Mechanisms of Action of FSH. Front Endocrinol (Lausanne) 2019; 10:593. [PMID: 31551928 PMCID: PMC6736554 DOI: 10.3389/fendo.2019.00593] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
|
29
|
Zhao P, Yu Y, Feng W, Du H, Yu J, Kang H, Zheng X, Wang Z, Liu GE, Ernst CW, Ran X, Wang J, Liu JF. Evidence of evolutionary history and selective sweeps in the genome of Meishan pig reveals its genetic and phenotypic characterization. Gigascience 2018; 7:5001425. [PMID: 29790964 PMCID: PMC6007440 DOI: 10.1093/gigascience/giy058] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 05/11/2018] [Indexed: 12/18/2022] Open
Abstract
Background Meishan is a pig breed indigenous to China and famous for its high fecundity. The traits of Meishan are strongly associated with its distinct evolutionary history and domestication. However, the genomic evidence linking the domestication of Meishan pigs with its unique features is still poorly understood. The goal of this study is to investigate the genomic signatures and evolutionary evidence related to the phenotypic traits of Meishan via large-scale sequencing. Results We found that the unique domestication of Meishan pigs occurred in the Taihu Basin area between the Majiabang and Liangzhu Cultures, during which 300 protein-coding genes have underwent positive selection. Notably, enrichment of the FoxO signaling pathway with significant enrichment signal and the harbored gene IGF1R were likely associated with the high fertility of Meishan pigs. Moreover, NFKB1 exhibited strong selective sweep signals and positively participated in hyaluronan biosynthesis as the key gene of NF-kB signaling, which may have resulted in the wrinkled skin and face of Meishan pigs. Particularly, three population-specific synonymous single-nucleotide variants occurred in PYROXD1, MC1R, and FAM83G genes; the T305C substitution in the MCIR gene explained the black coat of the Meishan pigs well. In addition, the shared haplotypes between Meishan and Duroc breeds confirmed the previous Asian-derived introgression and demonstrated the specific contribution of Meishan pigs. Conclusions These findings will help us explain the unique genetic and phenotypic characteristics of Meishan pigs and offer a plausible method for their utilization of Meishan pigs as valuable genetic resources in pig breeding and as an animal model for human wrinkled skin disease research.
Collapse
Affiliation(s)
- Pengju Zhao
- National Engineering Laboratory for Animal Breeding; Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ying Yu
- National Engineering Laboratory for Animal Breeding; Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wen Feng
- National Engineering Laboratory for Animal Breeding; Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Heng Du
- National Engineering Laboratory for Animal Breeding; Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jian Yu
- National Engineering Laboratory for Animal Breeding; Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Huimin Kang
- National Engineering Laboratory for Animal Breeding; Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xianrui Zheng
- National Engineering Laboratory for Animal Breeding; Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhiquan Wang
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, T6G 2C8, Canada
| | - George E Liu
- Animal Genomics and Improvement Laboratory, BARC, USDA-ARS, Beltsville, MD 20705-2350, USA
| | | | - Xueqin Ran
- School of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Jiafu Wang
- School of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Jian-Feng Liu
- National Engineering Laboratory for Animal Breeding; Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
30
|
Liu Y, Li S, Tao T, Li X, Zhu Q, Liao Y, Ma J, Sun Y, Liu W. Intrafollicular fibroblast growth factor 13 in polycystic ovary syndrome: relationship with androgen levels and oocyte developmental competence. J Ovarian Res 2018; 11:87. [PMID: 30257687 PMCID: PMC6158872 DOI: 10.1186/s13048-018-0455-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 09/04/2018] [Indexed: 01/14/2023] Open
Abstract
Background Fibroblast growth factor 13 (FGF13) is one of the most highly expressed FGF family members in adult mouse ovary. However, its precise roles in ovarian function remain largely unknown. We sought to evaluate the associations between FGF13 in follicular fluid and oocyte developmental competence in patients with polycystic ovary syndrome (PCOS). Methods A cross-sectional study was conducted on 43 patients with PCOS and 32 non-PCOS patients who underwent in vitro fertilization/intracytoplasmic sperm injection treatments. The highest quartiles of follicular fluid (FF)-FGF13 (≥117.51 pg/mL) and FF-total testosterone (FF-TT) (≥51.90 nmol/L) were defined as “elevated” FF-FGF13 levels and “elevated” FF-TT levels, respectively. Results The levels of FF-FGF13 were skewed, with a median of 82.97 pg/mL (59.79–117.51 pg/mL) in 75 patients. The prevalence of elevated FF-TT levels was significantly higher in the PCOS patients with elevated FF-FGF13 levels than in those without (64.3% vs. 35.7%, adjusted P = 0.0096). FF-TT and increased ovarian volume (> 10 mL for one or both ovaries) were positively correlated with FF-FGF13 in PCOS patients (r = 0.37, P = 0.013; r = 0.33, P = 0.032). A negative association was evident between FF-FGF13 and the MII oocyte rate in the multiple linear regression analysis (β = − 0.10, SE = 0.045, adjusted P = 0.027). However, the associations were not evident in the non-PCOS patients. Conclusions Our study suggests the presence of intrafollicular FGF13 in PCOS patients and implies that FGF13 might be involved in the pathophysiological process of PCOS. Electronic supplementary material The online version of this article (10.1186/s13048-018-0455-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Liu
- Department of Endocrinology, South Campus, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 201112, China
| | - Shengxian Li
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Tao Tao
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Xiaoxue Li
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Qinling Zhu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Yu Liao
- Department of Endocrinology, South Campus, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 201112, China
| | - Jing Ma
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Yun Sun
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200135, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| | - Wei Liu
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| |
Collapse
|
31
|
Cannarella R, Condorelli RA, La Vignera S, Calogero AE. Effects of the insulin-like growth factor system on testicular differentiation and function: a review of the literature. Andrology 2017; 6:3-9. [PMID: 29195026 DOI: 10.1111/andr.12444] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/21/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022]
Abstract
We recently described the occurrence of cryptorchidism, oligoasthenoteratozoospermia, and genital abnormalities in patients with distal 15q chromosome structural abnormalities. This observation brought us to hypothesize that insulin-like growth factor (IGF) receptor (IGF1R), mapping on the 15q 26.3 chromosomal band, may be involved in testicular function. To further evaluate this topic, we reviewed in vitro and in vivo studies exploring the role of the IGF system [IGF1, IGF2, IGF1R, insulin receptor substrates (IRS)] at the testicular level both in animals and in humans. In animals, IGF1/IGF1R has been found to be involved in testicular development during embryogenesis, in Sertoli cell (SC) proliferation, and in germ cell (GS) proliferation and differentiation. Interestingly, IGF1R seems to mediate follicle-stimulating hormone (FSH) effects through the PI3K/AKT pathway. In humans, IGF1 directly increases testicular volume. The molecular pathways responsible for testicular differentiation and IGF1/IGF1R signaling are highly conserved among species; therefore, the IGF system may be involved in FSH signaling also in humans. We suggest a possible molecular pathway occurring in human SCs, which involves both IGF1 and FSH through the PI3K/AKT pathway. The acknowledgment of an IGF1 mediation of the FSH-induced effects may open new ways for a targeted therapy in idiopathic non-FSH-responder oligoasthenoteratozoospermia.
Collapse
Affiliation(s)
- R Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - R A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - S La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - A E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
32
|
Law NC, Donaubauer EM, Zeleznik AJ, Hunzicker-Dunn M. How Protein Kinase A Activates Canonical Tyrosine Kinase Signaling Pathways To Promote Granulosa Cell Differentiation. Endocrinology 2017; 158:2043-2051. [PMID: 28460125 PMCID: PMC5505220 DOI: 10.1210/en.2017-00163] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/24/2017] [Indexed: 12/30/2022]
Abstract
Protein kinase A (PKA) has recently been shown to mimic the actions of follicle-stimulating hormone (FSH) by activating signaling pathways that promote granulosa cell (GC) differentiation, such as phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK). We sought to elucidate the mechanism by which PKA, a Ser/Thr kinase, intersected the PI3K/AKT and MAPK/ERK pathways that are canonically activated by receptor tyrosine kinases (RTKs). Our results show that for both of these pathways, the RTK is active in the absence of FSH yet signaling down the pathways to commence transcriptional responses requires FSH-stimulated PKA activation. For both pathways, PKA initiates signaling by regulating the activity of a protein phosphatase (PP). For the PI3K/AKT pathway, PKA activates the Ser/Thr PP1 complexed with the insulinlike growth factor 1 receptor (IGF-1R) and insulin receptor substrate 1 (IRS1) to dephosphorylate Ser residues on IRS1, authorizing phosphorylation of IRS1 by the IGF-1R to activate PI3K. Treatment of GCs with FSH and exogenous IGF-1 initiates synergistic IRS1 Tyr phosphorylation and resulting gene activation. The mechanism by which PKA activates PI3K is conserved in preovulatory GCs, MCF7 breast cancer cells, and FRTL thyroid cells. For the MAPK/ERK pathway, PKA promotes inactivation of the MAPK phosphatase (MKP) dual specificity phosphatase (DUSP) MKP3/DUSP6 to permit MEK-phosphorylated ERK to accumulate downstream of the epidermal growth factor receptor. Thus, for the two central signaling pathways that regulate gene expression in GCs, FSH via PKA intersects canonical RTK-regulated signaling by modulating the activity of PPs.
Collapse
Affiliation(s)
- Nathan C. Law
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164
| | - Elyse M. Donaubauer
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164
| | - Anthony J. Zeleznik
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee Women’s Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Mary Hunzicker-Dunn
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164
| |
Collapse
|
33
|
Baumgarten SC, Armouti M, Ko C, Stocco C. IGF1R Expression in Ovarian Granulosa Cells Is Essential for Steroidogenesis, Follicle Survival, and Fertility in Female Mice. Endocrinology 2017; 158:2309-2318. [PMID: 28407051 PMCID: PMC5505221 DOI: 10.1210/en.2017-00146] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/07/2017] [Indexed: 02/03/2023]
Abstract
Folliculogenesis is a lengthy process that requires the proliferation and differentiation of granulosa cells (GCs) for preovulatory follicle formation. The most crucial endocrine factor involved in this process is follicle-stimulating hormone (FSH). Interestingly, previous in vitro studies indicated that FSH does not stimulate GC proliferation in the absence of the insulinlike growth factor 1 receptor (IGF1R). To determine the role of the IGF1R in vivo, female mice with a conditional knockdown of the IGF1R in the GCs were produced and had undetectable levels of IGF1R mRNA and protein in the GCs. These animals were sterile, and their ovaries were smaller than those of control animals and contained no antral follicles even after gonadotropin stimulation. The lack of antral follicles correlated with a 90% decrease in serum estradiol levels. In addition, under a superovulation protocol no oocytes were found in the oviducts of these animals. Accordingly, the GCs of the mutant females expressed significantly lower levels of preovulatory markers including aromatase, luteinizing hormone receptor, and inhibin α. In contrast, no alterations in FSH receptor expression were observed in GCs lacking IGF1R. Immunohistochemistry studies demonstrated that ovaries lacking IGF1R had higher levels of apoptosis in follicles from the primary to the large secondary stages. Finally, molecular studies determined that protein kinase B activation was significantly impaired in mutant females when compared with controls. These in vivo findings demonstrate that IGF1R has a crucial role in GC function and, consequently, in female fertility.
Collapse
Affiliation(s)
- Sarah C. Baumgarten
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Marah Armouti
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - CheMyong Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana–Champaign, Champaigne, Illinois 60812
| | - Carlos Stocco
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| |
Collapse
|
34
|
Law NC, White MF, Hunzicker-Dunn ME. G protein-coupled receptors (GPCRs) That Signal via Protein Kinase A (PKA) Cross-talk at Insulin Receptor Substrate 1 (IRS1) to Activate the phosphatidylinositol 3-kinase (PI3K)/AKT Pathway. J Biol Chem 2016; 291:27160-27169. [PMID: 27856640 DOI: 10.1074/jbc.m116.763235] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/14/2016] [Indexed: 12/11/2022] Open
Abstract
G protein-coupled receptors (GPCRs) activate PI3K/v-AKT thymoma viral oncoprotein (AKT) to regulate many cellular functions that promote cell survival, proliferation, and growth. However, the mechanism by which GPCRs activate PI3K/AKT remains poorly understood. We used ovarian preantral granulosa cells (GCs) to elucidate the mechanism by which the GPCR agonist FSH via PKA activates the PI3K/AKT cascade. Insulin-like growth factor 1 (IGF1) is secreted in an autocrine/paracrine manner by GCs and activates the IGF1 receptor (IGF1R) but, in the absence of FSH, fails to stimulate YXXM phosphorylation of IRS1 (insulin receptor substrate 1) required for PI3K/AKT activation. We show that PKA directly phosphorylates the protein phosphatase 1 (PP1) regulatory subunit myosin phosphatase targeting subunit 1 (MYPT1) to activate PP1 associated with the IGF1R-IRS1 complex. Activated PP1 is sufficient to dephosphorylate at least four IRS1 Ser residues, Ser318, Ser346, Ser612, and Ser789, and promotes IRS1 YXXM phosphorylation by the IGF1R to activate the PI3K/AKT cascade. Additional experiments indicate that this mechanism also occurs in breast cancer, thyroid, and preovulatory granulosa cells, suggesting that the PKA-dependent dephosphorylation of IRS1 Ser/Thr residues is a conserved mechanism by which GPCRs signal to activate the PI3K/AKT pathway downstream of the IGF1R.
Collapse
Affiliation(s)
- Nathan C Law
- From the School of Molecular Biosciences, Washington State University, Pullman, Washington 99164 and
| | - Morris F White
- the Division of Endocrinology, Dept. of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Mary E Hunzicker-Dunn
- From the School of Molecular Biosciences, Washington State University, Pullman, Washington 99164 and
| |
Collapse
|
35
|
Stocco C. Follicle-stimulating Hormone (FSH) Phosphorylation of Protein Kinase B (AKT) Remains Controversial. J Biol Chem 2016; 291:14385. [PMID: 27371562 DOI: 10.1074/jbc.l116.726943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Carlos Stocco
- Department of Physiology and Biophysics, University of Illinois, Chicago, Illinois 60612.
| |
Collapse
|
36
|
Ulloa-Aguirre A, Zariñán T. The Follitropin Receptor: Matching Structure and Function. Mol Pharmacol 2016; 90:596-608. [PMID: 27382014 DOI: 10.1124/mol.116.104398] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/28/2016] [Indexed: 02/14/2025] Open
Abstract
Follitropin, or follicle-stimulating hormone (FSH) receptor (FSHR), is a G protein-coupled receptor belonging to the glycoprotein hormone receptor family that plays an essential role in reproduction. Although its primary location is the gonad, the FSHR has also been reported in extragonadal tissues including bone, placenta, endometrium, liver, and blood vessels from a number of malignant tumors. The recently resolved crystal structure of FSH bound to the entire FSHR ectodomain has been instrumental in more clearly defining the role of this domain in ligand binding and receptor activation. Biochemical, biophysical, and structural data also indicate that the FSHR exists as a higher order structure and that it may heterodimerize with its closely related receptor, the luteinizing hormone receptor; this association may have physiologic implications during ovarian follicle maturation given that both receptors may simultaneously coexist in the same cell. FSHR heterodimerization is unique to the ovary because in the testes, gonadotropin receptors are expressed in separate compartments. FSHR self-association appears to be required for receptor coupling to multiple effectors and adaptors, for the activation of multiple signaling pathways and the transduction of asymmetric signaling, and for negative and positive receptor cooperativity. It also provides a mechanism through which the glycosylation variants of FSH may exert distinct and differential effects at the target cell level. Given its importance in regulating activation of distinct signaling pathways, functional selectivity at the FSHR is briefly discussed, as well as the potential implications of this particular functional feature on the design of new pharmacological therapies in reproduction.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Research Support Network, National University of Mexico and National Institutes of Health, Mexico City, Mexico
| | - Teresa Zariñán
- Research Support Network, National University of Mexico and National Institutes of Health, Mexico City, Mexico
| |
Collapse
|
37
|
Herndon MK, Law NC, Donaubauer EM, Kyriss B, Hunzicker-Dunn M. Forkhead box O member FOXO1 regulates the majority of follicle-stimulating hormone responsive genes in ovarian granulosa cells. Mol Cell Endocrinol 2016; 434:116-26. [PMID: 27328024 PMCID: PMC4983523 DOI: 10.1016/j.mce.2016.06.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/26/2016] [Accepted: 06/16/2016] [Indexed: 01/11/2023]
Abstract
FSH promotes maturation of ovarian follicles. One pathway activated by FSH in granulosa cells (GCs) is phosphatidylinositol-3 kinase/AKT. The AKT target FOXO1 is reported to function primarily as a repressor of FSH genes, including Ccnd2 and Inha. Based on its broad functions in other tissues, we hypothesized that FOXO1 may regulate many more GC genes. We transduced GCs with empty adenovirus or constitutively active FOXO1 followed by treatment with FSH for 24 h, and conducted RNA deep sequencing. Results show that FSH regulates 3772 genes ≥2.0-fold; 60% of these genes are activated or repressed by FOXO1. Pathway Studio Analysis revealed enrichment of genes repressed by FOXO1 in metabolism, signaling, transport, development, and activated by FOXO1 in signaling, cytoskeletal functions, and apoptosis. Gene regulation was verified by q-PCR (eight genes) and ChIP analysis (two genes). We conclude that FOXO1 regulates the majority of FSH target genes in GCs.
Collapse
Affiliation(s)
- Maria K Herndon
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman WA 99163, USA.
| | - Nathan C Law
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman WA 99163, USA.
| | - Elyse M Donaubauer
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman WA 99163, USA.
| | - Brandon Kyriss
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman WA 99163, USA.
| | - Mary Hunzicker-Dunn
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman WA 99163, USA.
| |
Collapse
|
38
|
Hao M, Yuan F, Jin C, Zhou Z, Cao Q, Xu L, Wang G, Huang H, Yang D, Xie M, Zhao X. Overexpression of Lnk in the Ovaries Is Involved in Insulin Resistance in Women With Polycystic Ovary Syndrome. Endocrinology 2016; 157:3709-3718. [PMID: 27459314 PMCID: PMC5045500 DOI: 10.1210/en.2016-1234] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) progression involves abnormal insulin signaling. SH2 domain-containing adaptor protein (Lnk) may be an important regulator of the insulin signaling pathway. We investigated whether Lnk was involved in insulin resistance (IR). Thirty-seven women due to receive laparoscopic surgery from June 2011 to February 2012 were included from the gynecologic department of the Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University. Samples of polycystic and normal ovary tissues were examined by immunohistochemistry. Ovarian cell lines underwent insulin stimulation and Lnk overexpression. Expressed Lnk underwent coimmunoprecipitation tests with green fluorescent protein-labeled insulin receptor and His-tagged insulin receptor substrate 1 (IRS1), and their colocalization in HEK293T cells was examined. Ovarian tissues from PCOS patients with IR exhibited higher expression of Lnk than ovaries from normal control subjects and PCOS patients without IR; mainly in follicular granulosa cells, the follicular fluid and plasma of oocytes in secondary follicles, and atretic follicles. Lnk was coimmunoprecipitated with insulin receptor and IRS1. Lnk and insulin receptor/IRS1 locations overlapped around the nucleus. IR, protein kinase B (Akt), and ERK1/2 activities were inhibited by Lnk overexpression and inhibited further after insulin stimulation, whereas IRS1 serine activity was increased. Insulin receptor (Tyr1150/1151), Akt (Thr308), and ERK1/2 (Thr202/Tyr204) phosphorylation was decreased, whereas IRS1 (Ser307) phosphorylation was increased with Lnk overexpression. In conclusion, Lnk inhibits the phosphatidylinositol 3 kinase-AKT and MAPK-ERK signaling response to insulin. Higher expression of Lnk in PCOS suggests that Lnk probably plays a role in the development of IR.
Collapse
Affiliation(s)
- Meihua Hao
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Feng Yuan
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Chenchen Jin
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Zehong Zhou
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Qi Cao
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Ling Xu
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Guanlei Wang
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Hui Huang
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Dongzi Yang
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Meiqing Xie
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| | - Xiaomiao Zhao
- Department of Obstetrics and Gynecology (M.H., H.H., D.Y., M.X., X.Z.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Pharmacology (F.Y., C.J., G.W.), Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120 China; Department of Obstetrics and Gynecology (Z.Z.), Reproductive Medical Center, Peking University Third Hospital, Haidian District, Beijing, China; Division of Hematology/Oncology (Q.C.), Cedar-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California; and Cedar-Sinai Medical Center (L.X.), University of California, Los Angeles School of Medicine, Los Angeles, California
| |
Collapse
|
39
|
Donaubauer EM, Law NC, Hunzicker-Dunn ME. Follicle-Stimulating Hormone (FSH)-dependent Regulation of Extracellular Regulated Kinase (ERK) Phosphorylation by the Mitogen-activated Protein (MAP) Kinase Phosphatase MKP3. J Biol Chem 2016; 291:19701-12. [PMID: 27422819 DOI: 10.1074/jbc.m116.733972] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Indexed: 01/11/2023] Open
Abstract
Within the ovarian follicle, granulosa cells (GCs) surround and support immature oocytes. FSH promotes the differentiation and proliferation of GCs and is essential for fertility. We recently reported that ERK activation is necessary for FSH to induce key genes that define the preovulatory GC. This research focused on the phosphoregulation by FSH of ERK within GCs. FSH-stimulated ERK phosphorylation on Thr(202)/Tyr(204) was PKA-dependent, but MEK(Ser(217)/Ser(221)) phosphorylation was not regulated; rather, MEK was already active. However, treatment of GCs with the EGF receptor inhibitor AG1478, a dominant-negative RAS, an Src homology 2 domain-containing Tyr phosphatase inhibitor (NSC 87877), or the MEK inhibitor PD98059 blocked FSH-dependent ERK(Thr(202)/Tyr(204)) phosphorylation, demonstrating the requirement for upstream pathway components. We hypothesized that FSH via PKA enhances ERK phosphorylation by inhibiting the activity of a protein phosphatase that constitutively dephosphorylates ERK in the absence of FSH, allowing MEK-phosphorylated ERK to accumulate in the presence of FSH because of inactivation of the phosphatase. GCs treated with different phosphatase inhibitors permitted elimination of both Ser/Thr and Tyr phosphatases and implicated dual specificity phosphatases (DUSPs) in the dephosphorylation of ERK. Treatment with MAP kinase phosphatase (MKP3, DUSP6) inhibitors increased ERK(Thr(202)/Tyr(204)) phosphorylation in the absence of FSH to levels comparable with ERK phosphorylated in the presence of FSH. ERK co-immunoprecipitated with Myc-FLAG-tagged MKP3(DUSP6). GCs treated with MKP3(DUSP6) inhibitors blocked and PKA inhibitors enhanced dephosphorylation of recombinant ERK2-GST in an in vitro phosphatase assay. Together, these results suggest that FSH-stimulated ERK activation in GCs requires the PKA-dependent inactivation of MKP3(DUSP6).
Collapse
Affiliation(s)
- Elyse M Donaubauer
- From the School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Nathan C Law
- From the School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Mary E Hunzicker-Dunn
- From the School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| |
Collapse
|
40
|
Donaubauer EM, Hunzicker-Dunn ME. Extracellular Signal-regulated Kinase (ERK)-dependent Phosphorylation of Y-Box-binding Protein 1 (YB-1) Enhances Gene Expression in Granulosa Cells in Response to Follicle-stimulating Hormone (FSH). J Biol Chem 2016; 291:12145-60. [PMID: 27080258 DOI: 10.1074/jbc.m115.705368] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Indexed: 12/14/2022] Open
Abstract
Within the ovarian follicle, immature oocytes are surrounded and supported by granulosa cells (GCs). Stimulation of GCs by FSH leads to their proliferation and differentiation, events that are necessary for fertility. FSH activates multiple signaling pathways to regulate genes necessary for follicular maturation. Herein, we investigated the role of Y-box-binding protein-1 (YB-1) within GCs. YB-1 is a nucleic acid binding protein that regulates transcription and translation. Our results show that FSH promotes an increase in the phosphorylation of YB-1 on Ser(102) within 15 min that is maintained at significantly increased levels until ∼8 h post treatment. FSH-stimulated phosphorylation of YB-1(Ser(102)) is prevented by pretreatment of GCs with the PKA-selective inhibitor PKA inhibitor (PKI), the MEK inhibitor PD98059, or the ribosomal S6 kinase-2 (RSK-2) inhibitor BI-D1870. Thus, phosphorylation of YB-1 on Ser(102) is PKA-, ERK-, and RSK-2-dependent. However, pretreatment of GCs with the protein phosphatase 1 (PP1) inhibitor tautomycin increased phosphorylation of YB-1(Ser(102)) in the absence of FSH; FSH did not further increase YB-1(Ser(102)) phosphorylation. This result suggests that the major effect of RSK-2 is to inhibit PP1 rather than to directly phosphorylate YB-1 on Ser(102) YB-1 coimmunoprecipitated with PP1β catalytic subunit and RSK-2. Transduction of GCs with the dephospho-adenoviral-YB-1(S102A) mutant prevented the induction by FSH of Egfr, Cyp19a1, Inha, Lhcgr, Cyp11a1, Hsd17b1, and Pappa mRNAs and estradiol-17β production. Collectively, our results reveal that phosphorylation of YB-1 on Ser(102) via the ERK/RSK-2 signaling pathway is necessary for FSH-mediated expression of target genes required for maturation of follicles to a preovulatory phenotype.
Collapse
Affiliation(s)
- Elyse M Donaubauer
- From the School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Mary E Hunzicker-Dunn
- From the School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| |
Collapse
|