Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

Sphingosine-1-phosphate regulation of luteinising hormone-induced steroidogenesis and proliferation of bovine theca cells in vitro

Zaire B. Medina-Moctezuma A § , Cyndi G. Hernández-Coronado B § , Lydia Marín-López C , Adrián Guzmán B , David González-Aretia A , Carlos G. Gutiérrez D and Ana Ma. Rosales-Torres https://orcid.org/0000-0002-9077-3079 B *
+ Author Affiliations
- Author Affiliations

A Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México, México.

B Departamento Producción Agrícola y Animal, Universidad Autónoma Metropolitana-Xochimilco, Ciudad de México 04960, México.

C Maestría en Ciencias de la Producción y de la Salud Animal, Universidad Nacional Autónoma de México, Ciudad de México 04510, México.

D Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México 04510, México.


Handling Editor: Geraldine Hartshorne

Reproduction, Fertility and Development 35(9) 518-526 https://doi.org/10.1071/RD22289
Published online: 25 May 2023

© 2023 The Author(s) (or their employer(s)). Published by CSIRO Publishing

Abstract

Context: Sphingosine-1-phosphate (S1P) is synthesised by follicle granulosa cells under the influence of follicle-stimulating hormone and seems to be necessary for the biological effects of this gonadotrophin.

Aims: To determine if luteinising hormone (LH) increases S1P production and if this sphingolipid, either induced by LH or added to culture media, regulates steroidogenesis and cell viability in bovine theca cells.

Methods: We used bovine theca cell cultures treated with: S1P (0, 0.1, 1 and 10 μM; Experiment 1), LH (0, 0.02, 0.2 and 2 ng mL−1; Experiment 2) and LH (0.02 ng mL−1) plus a sphingosine kinase inhibitor (SKI-178; 0, 5 and 10 μM; Experiment 3).

Key results: Treatment with S1P did not affect (P > 0.05) theca cell viability or their ability to produce progesterone and testosterone. LH (0.02 ng mL−1) increased (P < 0.05) S1P production, and stimulated the expression of phosphorylated sphingosine kinase-1 (pSPHK1). However, the inhibition of SPHK1, by a specific SPHK1 inhibitor (SKI-178), reduced (P < 0.05) cell viability and progesterone secretion. Additionally, the use of SKI-178 increased theca cell testosterone production (P < 0.05).

Conclusions: S1P added to culture media did not affect cell viability or steroid synthesis. However, LH stimulated the production of S1P, by increasing phosphorylation of SPHK1 in theca cells. This intracellular S1P was inhibitory on testosterone production but augmented progesterone and viable cell number.

Implications: These results suggest a novel signalling pathway for LH in theca cells and underline the importance of S1P in the regulation of steroid synthesis.

Keywords: bovine follicles, cell viability, luteinising hormone, progesterone, sphingosine-1-phosphate, SPHK1, testosterone, theca cells.


References

Abedel-Majed, MA, Romereim, SM, Davis, JS, and Cupp, AS (2019). Perturbations in lineage specification of granulosa and theca cells may alter corpus luteum formation and function. Frontiers in Endocrinology 10, 832.
Perturbations in lineage specification of granulosa and theca cells may alter corpus luteum formation and function.Crossref | GoogleScholarGoogle Scholar |

Alvarez, SE, Harikumar, KB, Hait, NC, Allegood, J, Strub, GM, Kim, EY, Maceyka, M, Jiang, H, Luo, C, Kordula, T, Milstien, S, and Spiegel, S (2010). Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature 465, 1084–1088.
Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2.Crossref | GoogleScholarGoogle Scholar |

Arish, M, Alaidarous, M, Ali, R, Akhter, Y, and Rub, A (2017). Implication of sphingosine-1-phosphate signaling in diseases: molecular mechanism and therapeutic strategies. Journal of Receptors and Signal Transduction 37, 437–446.
Implication of sphingosine-1-phosphate signaling in diseases: molecular mechanism and therapeutic strategies.Crossref | GoogleScholarGoogle Scholar |

Becker, S, von Otte, S, Robenek, H, Diedrich, K, and Nofer, J-R (2011). Follicular fluid high-density lipoprotein-associated sphingosine 1-phosphate (S1P) promotes human granulosa lutein cell migration via S1P receptor type 3 and small G-protein RAC1. Biology of Reproduction 84, 604–612.
Follicular fluid high-density lipoprotein-associated sphingosine 1-phosphate (S1P) promotes human granulosa lutein cell migration via S1P receptor type 3 and small G-protein RAC1.Crossref | GoogleScholarGoogle Scholar |

Berndtson, AK, Vincent, SE, and Fortune, JE (1995). Low and high concentrations of gonadotropins differentially regulate hormone production by theca interna and granulosa cells from bovine preovulatory follicles. Biology of Reproduction 52, 1334–1342.

Brankin, V, Mitchell, MRP, Webb, B, and Hunter, MG (2003). Paracrine effects of oocyte secreted factors and stem cell factor on porcine granulosa and theca cells in vitro. Reproductive Biology and Endocrinology 1, 55.
Paracrine effects of oocyte secreted factors and stem cell factor on porcine granulosa and theca cells in vitro.Crossref | GoogleScholarGoogle Scholar |

Bravo, GÁ, Cedeño, RR, Casadevall, MP, and Ramió-Torrentà, L (2022). Sphingosine-1-Phosphate (S1P) and S1P signaling pathway modulators, from current insights to future perspectives. Cells 11, 2058.
Sphingosine-1-Phosphate (S1P) and S1P signaling pathway modulators, from current insights to future perspectives.Crossref | GoogleScholarGoogle Scholar |

Campbell, BK, Baird, DT, and Webb, R (1998). Effects of dose of LH on androgen production and luteinization of ovine theca cells cultured in a serum-free system. Reproduction 112, 69–77.
Effects of dose of LH on androgen production and luteinization of ovine theca cells cultured in a serum-free system.Crossref | GoogleScholarGoogle Scholar |

Campbell, BK, Kendall, NR, Onions, V, Guo, L, and Scaramuzzi, RJ (2014). Effect of monosaccharide sugars on LH-induced differentiation and sugar transport facilitator (SLC2A) expression in sheep theca cells in vitro. Reproduction, Fertility and Development 26, 453–461.
Effect of monosaccharide sugars on LH-induced differentiation and sugar transport facilitator (SLC2A) expression in sheep theca cells in vitro.Crossref | GoogleScholarGoogle Scholar |

Casarini, L, and Crépieux, P (2019). Molecular mechanisms of action of FSH. Frontiers in Endocrinology 10, 305.
Molecular mechanisms of action of FSH.Crossref | GoogleScholarGoogle Scholar |

Cheong, SH, Fortune, JE, Allen, JJ, Butler, WR, and Gilbert, RO (2020). Androgen production in response to LH is impaired in theca cells from nonovulatory dominant follicles in early-postpartum dairy cows. Domestic Animal Endocrinology 71, 106385.
Androgen production in response to LH is impaired in theca cells from nonovulatory dominant follicles in early-postpartum dairy cows.Crossref | GoogleScholarGoogle Scholar |

Chin, EC, and Abayasekara, DRE (2004). Progesterone secretion by luteinizing human granulosa cells: a possible cAMP-dependent but PKA-independent mechanism involved in its regulation. Journal of Endocrinology 183, 51–60.
Progesterone secretion by luteinizing human granulosa cells: a possible cAMP-dependent but PKA-independent mechanism involved in its regulation.Crossref | GoogleScholarGoogle Scholar |

Choi, H, and Roh, J (2020). LH-induced transcriptional regulation of KLF4 expression in granulosa cells occurs via the cAMP/PKA pathway and requires a putative Sp1 binding site. International Journal of Molecular Sciences 21, 7385.
LH-induced transcriptional regulation of KLF4 expression in granulosa cells occurs via the cAMP/PKA pathway and requires a putative Sp1 binding site.Crossref | GoogleScholarGoogle Scholar |

Clarke H (2017) Control of mammalian oocyte development by interactions with the maternal follicular environment. In ‘Oocytes’. (Ed. M Kloc) pp. 17–41. (Springer: Cham, Switzerland). https://doi.org/10.1007/978-3-319-60855-6_2

Dai, Y, Ivell, R, and Anand-Ivell, R (2017). Theca cell INSL3 and steroids together orchestrate the growing bovine antral follicle. Frontiers in Physiology 8, 1033.
Theca cell INSL3 and steroids together orchestrate the growing bovine antral follicle.Crossref | GoogleScholarGoogle Scholar |

Gupta, C, Chapekar, T, Chhabra, Y, Singh, P, Sinha, S, and Luthra, K (2012). Differential response to sustained stimulation by hCG & LH on goat ovarian granulosa cells. The Indian Journal of Medical Research 135, 331–340.

Hatzirodos, N, Glister, C, Hummitzsch, K, Irving-Rodgers, HF, Knight, PG, and Rodgers, RJ (2017). Transcriptomal profiling of bovine ovarian granulosa and theca interna cells in primary culture in comparison with their in vivo counterparts. PLoS ONE 12, e0173391.
Transcriptomal profiling of bovine ovarian granulosa and theca interna cells in primary culture in comparison with their in vivo counterparts.Crossref | GoogleScholarGoogle Scholar |

Hernández-Coronado, CG, Guzmán, A, Espinosa-Cervantes, R, Romano, MC, Verde-Calvo, JR, and Rosales-Torres, AM (2015). Sphingosine-1-phosphate and ceramide are associated with health and atresia of bovine ovarian antral follicles. Animal 9, 308–312.
Sphingosine-1-phosphate and ceramide are associated with health and atresia of bovine ovarian antral follicles.Crossref | GoogleScholarGoogle Scholar |

Hernández-Coronado, CG, Guzmán, A, Rodríguez, A, Mondragon, JA, Romano, MC, Gutiérrez, CG, and Rosales-Torres, AM (2016). Sphingosine-1-phosphate, regulated by FSH and VEGF, stimulates granulosa cell proliferation. General and Comparative Endocrinology 236, 1–8.
Sphingosine-1-phosphate, regulated by FSH and VEGF, stimulates granulosa cell proliferation.Crossref | GoogleScholarGoogle Scholar |

Hernández-Coronado, CG, Guzmán, A, Castillo-Juárez, H, Zamora-Gutiérrez, D, and Rosales-Torres, AM (2019). Sphingosine-1-phosphate (S1P) in ovarian physiology and disease. Annales d’Endocrinologie 80, 263–272.
Sphingosine-1-phosphate (S1P) in ovarian physiology and disease.Crossref | GoogleScholarGoogle Scholar |

Jiang, ZJ, Delaney, TL, Zanin, MP, Haberberger, RV, Pitson, SM, Huang, J, et al. (2019). Extracellular and intracellular sphingosine-1-phosphate distinctly regulates exocytosis in chromaffin cells. Journal of Neurochemistry 149, 729–746.
Extracellular and intracellular sphingosine-1-phosphate distinctly regulates exocytosis in chromaffin cells.Crossref | GoogleScholarGoogle Scholar |

Li, Y, Gao, D, Xu, T, Adur, MK, Zhang, L, Luo, L, Zhu, T, Tong, X, Zhang, D, Wang, Y, Ning, W, Qi, X, Cao, Z, and Zhang, Y (2020). Anti-Müllerian hormone inhibits luteinizing hormone-induced androstenedione synthesis in porcine theca cells. Theriogenology 142, 421–432.
Anti-Müllerian hormone inhibits luteinizing hormone-induced androstenedione synthesis in porcine theca cells.Crossref | GoogleScholarGoogle Scholar |

Lin, Z, Li, Y, Han, X, Fu, Z, Tian, Z, and Li, C (2022). Targeting SPHK1/PBX1 axis induced cell cycle arrest in non-small cell lung cancer. International Journal of Molecular Sciences 23, 12741.
Targeting SPHK1/PBX1 axis induced cell cycle arrest in non-small cell lung cancer.Crossref | GoogleScholarGoogle Scholar |

Liu, X, Qiao, P, Jiang, A, Jiang, J, Han, H, Wang, L, and Ren, C (2015). Paracrine regulation of steroidogenesis in theca cells by granulosa cells derived from mouse preantral follicles. BioMed Research International 2015, 925691.
Paracrine regulation of steroidogenesis in theca cells by granulosa cells derived from mouse preantral follicles.Crossref | GoogleScholarGoogle Scholar |

Lucki, NC, Li, D, and Sewer, MB (2012). Sphingosine-1-phosphate rapidly increases cortisol biosynthesis and the expression of genes involved in cholesterol uptake and transport in H295R adrenocortical cells. Molecular and Cellular Endocrinology 348, 165–175.
Sphingosine-1-phosphate rapidly increases cortisol biosynthesis and the expression of genes involved in cholesterol uptake and transport in H295R adrenocortical cells.Crossref | GoogleScholarGoogle Scholar |

Myat, TS, and Tetsuka, M (2017). Gossypol inhibits LH-induced steroidogenesis in bovine theca cells. Animal Science Journal 88, 63–71.
Gossypol inhibits LH-induced steroidogenesis in bovine theca cells.Crossref | GoogleScholarGoogle Scholar |

Nedresky D, Singh G (2022) ‘Physiology, luteinizing hormone.’ (StatPearls Publishing: Tampa, FL, USA)

Needle, E, Piparo, K, Cole, D, Worrall, C, Whitehead, I, Mahon, G, and Goldsmith, LT (2007). Protein kinase A-independent cAMP stimulation of progesterone in a luteal cell model is tyrosine kinase dependent but phosphatidylinositol-3-kinase and mitogen-activated protein kinase independent. Biology of Reproduction 77, 147–155.
Protein kinase A-independent cAMP stimulation of progesterone in a luteal cell model is tyrosine kinase dependent but phosphatidylinositol-3-kinase and mitogen-activated protein kinase independent.Crossref | GoogleScholarGoogle Scholar |

Nussbaum, C, Bannenberg, S, Keul, P, Gräler, MH, Gonçalves-de-Albuquerque, CF, Korhonen, H, von Wnuck Lipinski, K, Heusch, G, de Castro Faria Neto, HC, Rohwedder, I, Göthert, JR, Prasad, VP, Haufe, G, Lange-Sperandio, B, Offermanns, S, Sperandio, M, and Levkau, B (2015). Sphingosine-1-phosphate receptor 3 promotes leukocyte rolling by mobilizing endothelial P-selectin. Nature Communications 6, 6416.
Sphingosine-1-phosphate receptor 3 promotes leukocyte rolling by mobilizing endothelial P-selectin.Crossref | GoogleScholarGoogle Scholar |

Orisaka, M, Miyazaki, Y, Shirafuji, A, Tamamura, C, Tsuyoshi, H, Tsang, BK, and Yoshida, Y (2021). The role of pituitary gonadotropins and intraovarian regulators in follicle development: a mini-review. Reproductive Medicine and Biology 20, 169–175.
The role of pituitary gonadotropins and intraovarian regulators in follicle development: a mini-review.Crossref | GoogleScholarGoogle Scholar |

Paradiso, E, Lazzaretti, C, Sperduti, S, Antoniani, F, Fornari, G, Brigante, G, Di Rocco, G, Tagliavini, S, Trenti, T, Morini, D, Falbo, AI, Villani, MT, Nofer, J-R, Simoni, M, Potì, F, and Casarini, L (2021). Sphingosine-1 phosphate induces cAMP/PKA-independent phosphorylation of the cAMP response element-binding protein (CREB) in granulosa cells. Molecular and Cellular Endocrinology 520, 111082.
Sphingosine-1 phosphate induces cAMP/PKA-independent phosphorylation of the cAMP response element-binding protein (CREB) in granulosa cells.Crossref | GoogleScholarGoogle Scholar |

Pérez-Jeldres, T, Alvarez-Lobos, M, and Rivera-Nieves, J (2021). Targeting sphingosine-1-phosphate signaling in immune-mediated diseases: beyond multiple sclerosis. Drugs 81, 985–1002.
Targeting sphingosine-1-phosphate signaling in immune-mediated diseases: beyond multiple sclerosis.Crossref | GoogleScholarGoogle Scholar |

Qiao, H, Jiang, T, Mu, P, Chen, X, Wen, X, Hu, Z, Tang, S, Wen, J, and Deng, Y (2021). Cell fate determined by the activation balance between PKR and SPHK1. Cell Death & Differentiation 28, 401–418.
Cell fate determined by the activation balance between PKR and SPHK1.Crossref | GoogleScholarGoogle Scholar |

Rangel, PL, Rodríguez, A, Rojas, S, Sharp, PJ, and Gutierrez, CG (2009). Testosterone stimulates progesterone production and STAR, P450 cholesterol side-chain cleavage and LH receptor mRNAs expression in hen (Gallus domesticus) granulosa cells. Reproduction 138, 961–969.
Testosterone stimulates progesterone production and STAR, P450 cholesterol side-chain cleavage and LH receptor mRNAs expression in hen (Gallus domesticus) granulosa cells.Crossref | GoogleScholarGoogle Scholar |

Rosales-Torres, AM, Avalos-Rodríguez, A, Vergara-Onofre, M, Hernández-Pérez, O, Ballesteros, LM, García-Macedo, R, Ortiz-Navarrete, V, and Rosado, A (2000). Multiparametric study of atresia in ewe antral follicles: histology, flow cytometry, internucleosomal DNA fragmentation, and lysosomal enzyme activities in granulosa cells and follicular fluid. Molecular Reproduction and Development 55, 270–281.
Multiparametric study of atresia in ewe antral follicles: histology, flow cytometry, internucleosomal DNA fragmentation, and lysosomal enzyme activities in granulosa cells and follicular fluid.Crossref | GoogleScholarGoogle Scholar |

Rosales-Torres, AM, Guzmán-Sánchez, A, and Gutiérrez-Aguilar, C (2012). Follicular development in domestic ruminants. Tropical and Subtropical Agroecosystems 15, S147–S160.

Sarkar, S, Maceyka, M, Hait, NC, Paugh, SW, Sankala, H, Milstien, S, and Spiegel, S (2005). Sphingosine kinase 1 is required for migration, proliferation and survival of MCF-7 human breast cancer cells. FEBS Letters 579, 5313–5317.
Sphingosine kinase 1 is required for migration, proliferation and survival of MCF-7 human breast cancer cells.Crossref | GoogleScholarGoogle Scholar |

Shores, EM, Picton, HM, and Hunter, MG (2000). Differential regulation of pig theca cell steroidogenesis by LH, insulin-like growth factor I and granulosa cells in serum-free culture. Journal of Reproduction and Fertility 118, 211–219.
Differential regulation of pig theca cell steroidogenesis by LH, insulin-like growth factor I and granulosa cells in serum-free culture.Crossref | GoogleScholarGoogle Scholar |

Spiegel, S (2020). Sphingosine-1-phosphate: from insipid lipid to a key regulator. Journal of Biological Chemistry 295, 3371–3384.
Sphingosine-1-phosphate: from insipid lipid to a key regulator.Crossref | GoogleScholarGoogle Scholar |

Ulloa-Aguirre, A, Reiter, E, and Crépieux, P (2018). FSH receptor signaling: complexity of interactions and signal diversity. Endocrinology 159, 3020–3035.
FSH receptor signaling: complexity of interactions and signal diversity.Crossref | GoogleScholarGoogle Scholar |

Wang, P, Yuan, Y, Lin, W, Zhong, H, Xu, K, and Qi, X (2019). Roles of sphingosine-1-phosphate signaling in cancer. Cancer Cell International 19, 295.
Roles of sphingosine-1-phosphate signaling in cancer.Crossref | GoogleScholarGoogle Scholar |

Webb, R, Buratini, J, Hernández-Medrano, JH, Gutierrez, CG, and Campbell, BK (2016). Follicle development and selection: past, present and future. Animal Reproduction 13, 234–249.
Follicle development and selection: past, present and future.Crossref | GoogleScholarGoogle Scholar |

Weigert, A, Olesch, C, and Brüne, B (2019). Sphingosine-1-phosphate and macrophage biology – how the sphinx tames the big eater. Frontiers in Immunology 10, 1706.
Sphingosine-1-phosphate and macrophage biology – how the sphinx tames the big eater.Crossref | GoogleScholarGoogle Scholar |

Yin, J, Guo, Y-M, Chen, P, Xiao, H, Wang, X-H, Disanto, ME, and Zhang, X-H (2018). Testosterone regulates the expression and functional activity of sphingosine-1-phosphate receptors in the rat corpus cavernosum. Journal of Cellular and Molecular Medicine 22, 1507–1516.
Testosterone regulates the expression and functional activity of sphingosine-1-phosphate receptors in the rat corpus cavernosum.Crossref | GoogleScholarGoogle Scholar |