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

Transforming growth factor-β1 disrupts angiogenesis during the follicular–luteal transition through the Smad–serpin family E member 1 (SERPINE1)/serpin family B member 5 (SERPINB5) signalling pathway in the cow

Leyan Yan https://orcid.org/0000-0003-1562-2550 A B , Xiaolu Qu A B , Jianning Yu A B , Robert S. Robinson C , Kathryn J. Woad C and Zhendan Shi A B D
+ Author Affiliations
- Author Affiliations

A Laboratory of Animal Improvement and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.

B Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.

C School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire LE12 5RD, UK.

D Corresponding author. Email: zdshi@jaas.ac.cn

Reproduction, Fertility and Development 33(10) 643-654 https://doi.org/10.1071/RD20325
Submitted: 14 December 2020  Accepted: 13 April 2021   Published: 19 May 2021

Abstract

Intense angiogenesis is critical for the development of the corpus luteum and is tightly regulated by numerous factors. However, the exact role transforming growth factor-β1 (TGFB1) plays during this follicular–luteal transition remains unclear. This study hypothesised that TGFB1, acting through TGFB receptor 1 (TGFBR1) and Smad2/3 signalling, would suppress angiogenesis during the follicular–luteal transition. Using a serum-free luteinising follicular angiogenesis culture system, TGFB1 (1 and 10 ng mL–1) markedly disrupted the formation of capillary-like structures, reducing the endothelial cell network area and the number of branch points (P < 0.001 compared with control). Furthermore, TGFB1 activated canonical Smad signalling and inhibited endothelial nitric oxide synthase (NOS3) mRNA expression, but upregulated latent TGFB-binding protein and TGFBR1, serpin family E member 1 (SERPINE1) and serpin family B member 5 (SERPINB5) mRNA expression. SB431542, a TGFBR1 inhibitor, reversed the TGFB1-induced upregulation of SERPINE1 and SERPINB5. In addition, TGFB1 reduced progesterone synthesis by decreasing the expression of steroidogenic acute regulatory protein (STAR), cytochrome P450 family 11 subfamily A member 1 (CYP11A1) and 3β-hydroxysteroid dehydrogenase (HSD3B1) expression. These results show that TGFB1 regulates NOS3, SERPINE1 and SERPINB5 expression via TGFBR1 and Smad2/3 signalling and this could be the mechanism by which TGFB1 suppresses endothelial networks. Thereby, TGFB1 may provide critical homeostatic control of angiogenesis during the follicular–luteal transition. The findings of this study reveal the molecular mechanisms underlying the actions of TGFB1 in early luteinisation, which may lead to novel therapeutic strategies to reverse luteal inadequacy.

Keywords: angiogenesis, luteinisation, serpin family B member 5 (SERPINB5), serpin family E member 1 (SERPINE1), Smad, transforming growth factor-β1 (TGFB1).


References

Andersen, C. L., Jensen, J. L., and Ørntoft, T. F. (2004). Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res. 64, 5245–5250.
Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets.Crossref | GoogleScholarGoogle Scholar | 15289330PubMed |

Berisha, B., Steffl, M., Welter, H., Kliem, H., Meyer, H. H., Schams, D., and Amselgruber, W. (2008). Effect of the luteinising hormone surge on regulation of vascular endothelial growth factor and extracellular matrix-degrading proteinases and their inhibitors in bovine follicles. Reprod. Fertil. Dev. 20, 258–268.
Effect of the luteinising hormone surge on regulation of vascular endothelial growth factor and extracellular matrix-degrading proteinases and their inhibitors in bovine follicles.Crossref | GoogleScholarGoogle Scholar | 18255015PubMed |

Bodenstine, T. M., Seftor, R. E., Khalkhali-Ellis, Z., Seftor, E. A., Pemberton, P. A., and Hendrix, M. J. (2012). Maspin: molecular mechanisms and therapeutic implications. Cancer Metastasis Rev. 31, 529–551.
Maspin: molecular mechanisms and therapeutic implications.Crossref | GoogleScholarGoogle Scholar | 22752408PubMed |

Boehm, J. R., Kutz, S. M., Sage, E. H., Staiano-Coico, L., and Higgins, P. J. (1999). Growth state-dependent regulation of plasminogen activator inhibitor type-1 gene expression during epithelial cell stimulation by serum and transforming growth factor-beta1. J. Cell. Physiol. 181, 96–106.
Growth state-dependent regulation of plasminogen activator inhibitor type-1 gene expression during epithelial cell stimulation by serum and transforming growth factor-beta1.Crossref | GoogleScholarGoogle Scholar | 10457357PubMed |

Dodson, W. C., and Schomberg, D. W. (1987). The Effect of Transforming Growth Factor-β on Follicle-Stimulating Hormone-Induced Differentiation of Cultured Rat Granulosa Cells. Endocrinology 120, 512–516.
The Effect of Transforming Growth Factor-β on Follicle-Stimulating Hormone-Induced Differentiation of Cultured Rat Granulosa Cells.Crossref | GoogleScholarGoogle Scholar | 3026778PubMed |

Fang, L., Chang, H. M., Cheng, J. C., Leung, P. C., and Sun, Y. P. (2014). TGF-β1 downregulates StAR expression and decreases progesterone production through Smad3 and ERK1/2 signaling pathways in human granulosa cells. J. Clin. Endocrinol. Metab. 99, E2234–E2243.
TGF-β1 downregulates StAR expression and decreases progesterone production through Smad3 and ERK1/2 signaling pathways in human granulosa cells.Crossref | GoogleScholarGoogle Scholar | 25140399PubMed |

Farberov, S., and Meidan, R. (2016). Thrombospondin-1 Affects Bovine Luteal Function via Transforming Growth Factor-Beta1-Dependent and Independent Actions. Biol. Reprod. 94, 25.
Thrombospondin-1 Affects Bovine Luteal Function via Transforming Growth Factor-Beta1-Dependent and Independent Actions.Crossref | GoogleScholarGoogle Scholar | 26658711PubMed |

Fazzini, M., Vallejo, G., Colman-Lerner, A., Trigo, R., Campo, S., Barañao, J. L., and Saragüeta, P. E. (2006). Transforming growth factor beta1 regulates follistatin mRNA expression during in vitro bovine granulosa cell differentiation. J. Cell. Physiol. 207, 40–48.
Transforming growth factor beta1 regulates follistatin mRNA expression during in vitro bovine granulosa cell differentiation.Crossref | GoogleScholarGoogle Scholar | 16245315PubMed |

Ferrari, G., Pintucci, G., Seghezzi, G., Hyman, K., Galloway, A. C., and Mignatti, P. (2006). VEGF, a prosurvival factor, acts in concert with TGF-beta1 to induce endothelial cell apoptosis. Proc. Natl. Acad. Sci. USA 103, 17260–17265.
VEGF, a prosurvival factor, acts in concert with TGF-beta1 to induce endothelial cell apoptosis.Crossref | GoogleScholarGoogle Scholar | 17088559PubMed |

Fraser, H. M., Bell, J., Wilson, H., Taylor, P. D., Morgan, K., Anderson, R. A., and Duncan, W. C. (2005). Localization and quantification of cyclic changes in the expression of endocrine gland vascular endothelial growth factor in the human corpus luteum. J. Clin. Endocrinol. Metab. 90, 427–434.
Localization and quantification of cyclic changes in the expression of endocrine gland vascular endothelial growth factor in the human corpus luteum.Crossref | GoogleScholarGoogle Scholar | 15483093PubMed |

Fraser, H. M., Hastings, J. M., Allan, D., Morris, K. D., Rudge, J. S., and Wiegand, S. J. (2012). Inhibition of delta-like ligand 4 induces luteal hypervascularization followed by functional and structural luteolysis in the primate ovary. Endocrinology 153, 1972–1983.
Inhibition of delta-like ligand 4 induces luteal hypervascularization followed by functional and structural luteolysis in the primate ovary.Crossref | GoogleScholarGoogle Scholar | 22334711PubMed |

Gangrade, B. K., Gotcher, E. D., Davis, J. S., and May, J. V. (1993). The secretion of transforming growth factor-beta by bovine luteal cells in vitro. Mol. Cell. Endocrinol. 93, 117–123.
The secretion of transforming growth factor-beta by bovine luteal cells in vitro.Crossref | GoogleScholarGoogle Scholar | 8349022PubMed |

Ghiglieri, C., Khatchadourian, C., Tabone, E., Hendrick, J. C., Benahmed, M., and Ménézo, Y. (1995). Immunolocalization of transforming growth factor-beta 1 and transforming growth factor-beta 2 in the mouse ovary during gonadotrophin-induced follicular maturation. Hum. Reprod. 10, 2115–2119.
Immunolocalization of transforming growth factor-beta 1 and transforming growth factor-beta 2 in the mouse ovary during gonadotrophin-induced follicular maturation.Crossref | GoogleScholarGoogle Scholar | 8567851PubMed |

Goumans, M. J., and Ten Dijke, P. (2018). TGF-β Signaling in Control of Cardiovascular Function. Cold Spring Harb. Perspect. Biol. 10, a022210.
TGF-β Signaling in Control of Cardiovascular Function.Crossref | GoogleScholarGoogle Scholar | 28348038PubMed |

Goumans, M. J., Lebrin, F., and Valdimarsdottir, G. (2003). Controlling the angiogenic switch: a balance between two distinct TGF-b receptor signaling pathways. Trends Cardiovasc. Med. 13, 301–307.
Controlling the angiogenic switch: a balance between two distinct TGF-b receptor signaling pathways.Crossref | GoogleScholarGoogle Scholar | 14522471PubMed |

Goumans, M. J., Liu, Z., and ten Dijke, P. (2009). TGF-beta signaling in vascular biology and dysfunction. Cell Res. 19, 116–127.
TGF-beta signaling in vascular biology and dysfunction.Crossref | GoogleScholarGoogle Scholar | 19114994PubMed |

Hayashi, K. G., Ushizawa, K., Hosoe, M., and Takahashi, T. (2011). Differential gene expression of serine protease inhibitors in bovine ovarian follicle: possible involvement in follicular growth and atresia. Reprod. Biol. Endocrinol. 9, 72.
Differential gene expression of serine protease inhibitors in bovine ovarian follicle: possible involvement in follicular growth and atresia.Crossref | GoogleScholarGoogle Scholar | 21619581PubMed |

Heldin, C. H., Miyazono, K., and ten Dijke, P. (1997). TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature 390, 465–471.
TGF-beta signalling from cell membrane to nucleus through SMAD proteins.Crossref | GoogleScholarGoogle Scholar | 9393997PubMed |

Henkes, L. E., Sullivan, B. T., Lynch, M. P., Kolesnick, R., Arsenault, D., Puder, M., Davis, J. S., and Rueda, B. R. (2008). Acid sphingomyelinase involvement in tumor necrosis factor alpha-regulated vascular and steroid disruption during luteolysis in vivo. Proc. Natl. Acad. Sci. USA 105, 7670–7675.
Acid sphingomyelinase involvement in tumor necrosis factor alpha-regulated vascular and steroid disruption during luteolysis in vivo.Crossref | GoogleScholarGoogle Scholar | 18505843PubMed |

Hou, X., Arvisais, E. W., Jiang, C., Chen, D. B., Roy, S. K., Pate, J. L., Hansen, T. R., Rueda, B. R., and Davis, J. S. (2008). Prostaglandin F2alpha stimulates the expression and secretion of transforming growth factor B1 via induction of the early growth response 1 gene (EGR1) in the bovine corpus luteum. Mol. Endocrinol. 22, 403–414.
Prostaglandin F2alpha stimulates the expression and secretion of transforming growth factor B1 via induction of the early growth response 1 gene (EGR1) in the bovine corpus luteum.Crossref | GoogleScholarGoogle Scholar | 17916653PubMed |

Inman, G. J., Nicolás, F. J., Callahan, J. F., Harling, J. D., Gaster, L. M., Reith, A. D., Laping, N. J., and Hill, C. S. (2002). SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. Mol. Pharmacol. 62, 65–74.
SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7.Crossref | GoogleScholarGoogle Scholar | 12065756PubMed |

Inoue, N., Venema, R. C., Sayegh, H. S., Ohara, Y., Murphy, T. J., and Harrison, D. G. (1995). Molecular regulation of the bovine endothelial cell nitric oxide synthase by transforming growth factor-beta 1. Arterioscler. Thromb. Vasc. Biol. 15, 1255–1261.
Molecular regulation of the bovine endothelial cell nitric oxide synthase by transforming growth factor-beta 1.Crossref | GoogleScholarGoogle Scholar | 7543000PubMed |

Jakobsson, L., and van Meeteren, L. A. (2013). Transforming growth factor β family members in regulation of vascular function: in the light of vascular conditional knockouts. Exp. Cell Res. 319, 1264–1270.
Transforming growth factor β family members in regulation of vascular function: in the light of vascular conditional knockouts.Crossref | GoogleScholarGoogle Scholar | 23454603PubMed |

Jarad, M., Kuczynski, E. A., Morrison, J., Viloria-Petit, A. M., and Coomber, B. L. (2017). Release of endothelial cell associated VEGFR2 during TGF-β modulated angiogenesis in vitro. BMC Cell Biol. 18, 10.
Release of endothelial cell associated VEGFR2 during TGF-β modulated angiogenesis in vitro.Crossref | GoogleScholarGoogle Scholar | 28114883PubMed |

Jayawardana, B. C., Shimizu, T., Nishimoto, H., Kaneko, E., Tetsuka, M., and Miyamoto, A. (2006). Hormonal regulation of expression of growth differentiation factor-9 receptor type I and II genes in the bovine ovarian follicle. Reproduction 131, 545–553.
Hormonal regulation of expression of growth differentiation factor-9 receptor type I and II genes in the bovine ovarian follicle.Crossref | GoogleScholarGoogle Scholar | 16514197PubMed |

Joseph, C., Hunter, M. G., Sinclair, K. D., and Robinson, R. S. (2012). The expression, regulation and function of secreted protein, acidic, cysteine-rich in the follicle-luteal transition. Reproduction 144, 361–372.
The expression, regulation and function of secreted protein, acidic, cysteine-rich in the follicle-luteal transition.Crossref | GoogleScholarGoogle Scholar | 22733805PubMed |

Kliem, H., Welter, H., Kraetzl, W. D., Steffl, M., Meyer, H. H., Schams, D., and Berisha, B. (2007). Expression and localisation of extracellular matrix degrading proteases and their inhibitors during the oestrous cycle and after induced luteolysis in the bovine corpus luteum. Reproduction 134, 535–547.
Expression and localisation of extracellular matrix degrading proteases and their inhibitors during the oestrous cycle and after induced luteolysis in the bovine corpus luteum.Crossref | GoogleScholarGoogle Scholar | 17709571PubMed |

Knight, P. G., and Glister, C. (2006). TGF-beta superfamily members and ovarian follicle development. Reproduction 132, 191–206.
TGF-beta superfamily members and ovarian follicle development.Crossref | GoogleScholarGoogle Scholar | 16885529PubMed |

Kubota, T., Kamada, S., Taguchi, M., and Aso, T. (1994). Fertilization and early embryology: Autocrine/paracrine function of transforming growth factor-β1 in porcine granulosa cells. Hum. Reprod. 9, 2118–2122.
Fertilization and early embryology: Autocrine/paracrine function of transforming growth factor-β1 in porcine granulosa cells.Crossref | GoogleScholarGoogle Scholar | 7868683PubMed |

Kwak, J. H., Woo, J. S., Shin, K., Kim, H. J., Jeong, H. S., Han, D. C., Kim, S. I., and Park, C. S. (2005). Expression and regulation of latent TGF-beta binding protein-1 transcripts and their splice variants in human glomerular endothelial cells. J. Korean Med. Sci. 20, 628–635.
Expression and regulation of latent TGF-beta binding protein-1 transcripts and their splice variants in human glomerular endothelial cells.Crossref | GoogleScholarGoogle Scholar | 16100456PubMed |

Laird, M., Woad, K. J., Hunter, M. G., Mann, G. E., and Robinson, R. S. (2013). Fibroblast growth factor 2 induces the precocious development of endothelial cell networks in bovine luteinising follicular cells. Reprod. Fertil. Dev. 25, 372–386.
Fibroblast growth factor 2 induces the precocious development of endothelial cell networks in bovine luteinising follicular cells.Crossref | GoogleScholarGoogle Scholar | 23153420PubMed |

Lebrin, F., Deckers, M., Bertolino, P., and Ten Dijke, P. (2005). TGF-beta receptor function in the endothelium. Cardiovasc. Res. 65, 599–608.
TGF-beta receptor function in the endothelium.Crossref | GoogleScholarGoogle Scholar | 15664386PubMed |

LeCouter, J., Lin, R., and Ferrara, N. (2002). Endocrine gland-derived VEGF and the emerging hypothesis of organ-specific regulation of angiogenesis. Nat. Med. 8, 913–917.
Endocrine gland-derived VEGF and the emerging hypothesis of organ-specific regulation of angiogenesis.Crossref | GoogleScholarGoogle Scholar | 12205443PubMed |

Livak, K. J., and Schmittgen, T. D. (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402–408.
Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method.Crossref | GoogleScholarGoogle Scholar | 11846609PubMed |

Ma, Z., Guo, J., Zhang, Y., Zhang, Y., Zhang, M., Zong, R., Chen, F., and Zhang, J. (2020). Neuromedin B regulates steroidogenesis, cell viability and apoptosis in rabbit Leydig cells. Gen. Comp. Endocrinol. 288, 113371.
Neuromedin B regulates steroidogenesis, cell viability and apoptosis in rabbit Leydig cells.Crossref | GoogleScholarGoogle Scholar | 31857076PubMed |

Maroni, D., and Davis, J. S. (2011). TGFB1 disrupts the angiogenic potential of microvascular endothelial cells of the corpus luteum. J. Cell Sci. 124, 2501–2510.
TGFB1 disrupts the angiogenic potential of microvascular endothelial cells of the corpus luteum.Crossref | GoogleScholarGoogle Scholar | 21693577PubMed |

Martelli, A., Berardinelli, P., Russo, V., Mauro, A., Bernabò, N., Gioia, L., Mattioli, M., and Barboni, B. (2006). Spatio-temporal analysis of vascular endothelial growth factor expression and blood vessel remodelling in pig ovarian follicles during the periovulatory period. J. Mol. Endocrinol. 36, 107–119.
Spatio-temporal analysis of vascular endothelial growth factor expression and blood vessel remodelling in pig ovarian follicles during the periovulatory period.Crossref | GoogleScholarGoogle Scholar | 16461931PubMed |

Mattar, D., Samir, M., Laird, M., and Knight, P. G. (2020). Modulatory effects of TGF-β1 and BMP6 on thecal angiogenesis and steroidogenesis in the bovine ovary. Reproduction 159, 397–408.
Modulatory effects of TGF-β1 and BMP6 on thecal angiogenesis and steroidogenesis in the bovine ovary.Crossref | GoogleScholarGoogle Scholar | 31967968PubMed |

McCann, J. V., Xiao, L., Kim, D. J., Khan, O. F., Kowalski, P. S., Anderson, D. G., Pecot, C. V., Azam, S. H., Parker, J. S., Tsai, Y. S., Wolberg, A. S., Turner, S. D., Tatsumi, K., Mackman, N., and Dudley, A. C. (2019). Endothelial miR-30c suppresses tumor growth via inhibition of TGF-β-induced Serpine1. J. Clin. Invest. 129, 1654–1670.
Endothelial miR-30c suppresses tumor growth via inhibition of TGF-β-induced Serpine1.Crossref | GoogleScholarGoogle Scholar | 30855280PubMed |

Nilsson, E. E., Doraiswamy, V., and Skinner, M. K. (2003). Transforming growth factor-beta isoform expression during bovine ovarian antral follicle development. Mol. Reprod. Dev. 66, 237–246.
Transforming growth factor-beta isoform expression during bovine ovarian antral follicle development.Crossref | GoogleScholarGoogle Scholar | 14502602PubMed |

Öklü, R., and Hesketh, R. (2000). The latent transforming growth factor beta binding protein (LTBP) family. Biochem. J. 352, 601–610.
The latent transforming growth factor beta binding protein (LTBP) family.Crossref | GoogleScholarGoogle Scholar | 11104663PubMed |

Payne, A. H., and Hales, D. B. (2004). Overview of steroidogenic enzymes in the pathway from cholesterol to active steroid hormones. Endocr. Rev. 25, 947–970.
Overview of steroidogenic enzymes in the pathway from cholesterol to active steroid hormones.Crossref | GoogleScholarGoogle Scholar | 15583024PubMed |

Pepper, M. S. (1997). Transforming growth factor-beta: vasculogenesis, angiogenesis, and vessel wall integrity. Cytokine Growth Factor Rev. 8, 21–43.
Transforming growth factor-beta: vasculogenesis, angiogenesis, and vessel wall integrity.Crossref | GoogleScholarGoogle Scholar | 9174661PubMed |

Pepper, M. S., Belin, D., Montesano, R., Orci, L., and Vassalli, J. D. (1990). Transforming growth factor-beta 1 modulates basic fibroblast growth factor-induced proteolytic and angiogenic properties of endothelial cells in vitro. J. Cell Biol. 111, 743–755.
Transforming growth factor-beta 1 modulates basic fibroblast growth factor-induced proteolytic and angiogenic properties of endothelial cells in vitro.Crossref | GoogleScholarGoogle Scholar | 1696269PubMed |

Peshavariya, H. M., Chan, E. C., Liu, G. S., Jiang, F., and Dusting, G. J. (2014). Transforming growth factor-β1 requires NADPH oxidase 4 for angiogenesis in vitro and in vivo. J. Cell. Mol. Med. 18, 1172–1183.
Transforming growth factor-β1 requires NADPH oxidase 4 for angiogenesis in vitro and in vivo.Crossref | GoogleScholarGoogle Scholar | 24629065PubMed |

Qin, L., and Zhang, M. (2010). Maspin regulates endothelial cell adhesion and migration through an integrin signaling pathway. J. Biol. Chem. 285, 32360–32369.
Maspin regulates endothelial cell adhesion and migration through an integrin signaling pathway.Crossref | GoogleScholarGoogle Scholar | 20713357PubMed |

Qiu, F., Tong, H., Wang, Y., Tao, J., Wang, H., and Chen, L. (2018). Recombinant human maspin inhibits high glucose-induced oxidative stress and angiogenesis of human retinal microvascular endothelial cells via PI3K/AKT pathway. Mol. Cell. Biochem. 446, 127–136.
Recombinant human maspin inhibits high glucose-induced oxidative stress and angiogenesis of human retinal microvascular endothelial cells via PI3K/AKT pathway.Crossref | GoogleScholarGoogle Scholar | 29363056PubMed |

Qu, X., Yan, L., Guo, R., Li, H., and Shi, Z. (2019). ROS-Induced GATA4 and GATA6 Downregulation Inhibits StAR Expression in LPS-Treated Porcine Granulosa-Lutein Cells. Oxid. Med. Cell. Longev. 2019, 5432792.
ROS-Induced GATA4 and GATA6 Downregulation Inhibits StAR Expression in LPS-Treated Porcine Granulosa-Lutein Cells.Crossref | GoogleScholarGoogle Scholar | 31178965PubMed |

Reynolds, L. P., Grazul-Bilska, A. T., and Redmer, D. A. (2000). Angiogenesis in the corpus luteum. Endocrine 12, 1–9.
Angiogenesis in the corpus luteum.Crossref | GoogleScholarGoogle Scholar | 10855683PubMed |

Robertson, I. B., Horiguchi, M., Zilberberg, L., Dabovic, B., Hadjiolova, K., and Rifkin, D. B. (2015). Latent TGF-β-binding proteins. Matrix Biol. 47, 44–53.
Latent TGF-β-binding proteins.Crossref | GoogleScholarGoogle Scholar | 25960419PubMed |

Robinson, R. S., Hammond, A. J., Mann, G. E., and Hunter, M. G. (2008). A novel physiological culture system that mimics luteal angiogenesis. Reproduction 135, 405–413.
A novel physiological culture system that mimics luteal angiogenesis.Crossref | GoogleScholarGoogle Scholar | 18299434PubMed |

Robinson, R. S., Woad, K. J., Hammond, A. J., Laird, M., Hunter, M. G., and Mann, G. E. (2009). Angiogenesis and vascular function in the ovary. Reproduction 138, 869–881.
Angiogenesis and vascular function in the ovary.Crossref | GoogleScholarGoogle Scholar | 19786399PubMed |

Roelen, B. A., Van Eijk, M. J., Van Rooijen, M. A., Bevers, M. M., Larson, J. H., Lewin, H. A., and Mummery, C. L. (1998). Molecular cloning, genetic mapping, and developmental expression of a bovine transforming growth factor beta (TGF-beta) type I receptor. Mol. Reprod. Dev. 49, 1–9.
Molecular cloning, genetic mapping, and developmental expression of a bovine transforming growth factor beta (TGF-beta) type I receptor.Crossref | GoogleScholarGoogle Scholar | 9406190PubMed |

Sadler, J. E. (1991). von Willebrand factor. J. Biol. Chem. 266, 22777–22780.
von Willebrand factor.Crossref | GoogleScholarGoogle Scholar | 1744071PubMed |

Santibanez, J. F., Letamendia, A., Perez-Barriocanal, F., Silvestri, C., Saura, M., Vary, C. P., Lopez-Novoa, J. M., Attisano, L., and Bernabeu, C. (2007). Endoglin increases eNOS expression by modulating Smad2 protein levels and Smad2-dependent TGF-beta signaling. J. Cell. Physiol. 210, 456–468.
Endoglin increases eNOS expression by modulating Smad2 protein levels and Smad2-dependent TGF-beta signaling.Crossref | GoogleScholarGoogle Scholar | 17058229PubMed |

Saura, M., Zaragoza, C., Cao, W., Bao, C., Rodríguez-Puyol, M., Rodríguez-Puyol, D., and Lowenstein, C. J. (2002). Smad2 mediates transforming growth factor-beta induction of endothelial nitric oxide synthase expression. Circ. Res. 91, 806–813.
Smad2 mediates transforming growth factor-beta induction of endothelial nitric oxide synthase expression.Crossref | GoogleScholarGoogle Scholar | 12411395PubMed |

Serratì, S., Margheri, F., Pucci, M., Cantelmo, A. R., Cammarota, R., Dotor, J., Borràs-Cuesta, F., Fibbi, G., Albini, A., and Del Rosso, M. (2009). TGFbeta1 antagonistic peptides inhibit TGFbeta1-dependent angiogenesis. Biochem. Pharmacol. 77, 813–825.
TGFbeta1 antagonistic peptides inhibit TGFbeta1-dependent angiogenesis.Crossref | GoogleScholarGoogle Scholar | 19041849PubMed |

Sessa, W. C. (2004). eNOS at a glance. J. Cell Sci. 117, 2427–2429.
eNOS at a glance.Crossref | GoogleScholarGoogle Scholar | 15159447PubMed |

Skinner, M. K., Keski-Oja, J., Osteen, K. G., and Moses, H. L. (1987). Ovarian thecal cells produce transforming growth factor-beta which can regulate granulosa cell growth. Endocrinology 121, 786–792.
Ovarian thecal cells produce transforming growth factor-beta which can regulate granulosa cell growth.Crossref | GoogleScholarGoogle Scholar | 3297652PubMed |

Smith, M. F., McIntush, E. W., Ricke, W. A., Kojima, F. N., and Smith, G. W. (1999). Regulation of ovarian extracellular matrix remodelling by metalloproteinases and their tissue inhibitors: effects on follicular development, ovulation and luteal function. J. Reprod. Fertil. Suppl. 54, 367–381.
| 10692869PubMed |

Sriperumbudur, R., Zorrilla, L., and Gadsby, J. E. (2010). Transforming growth factor-β (TGFβ) and its signaling components in peri-ovulatory pig follicles. Anim. Reprod. Sci. 120, 84–94.
Transforming growth factor-β (TGFβ) and its signaling components in peri-ovulatory pig follicles.Crossref | GoogleScholarGoogle Scholar | 20378284PubMed |

Stocco, C., Telleria, C., and Gibori, G. (2007). The molecular control of corpus luteum formation, function, and regression. Endocr. Rev. 28, 117–149.
The molecular control of corpus luteum formation, function, and regression.Crossref | GoogleScholarGoogle Scholar | 17077191PubMed |

ten Dijke, P., and Arthur, H. M. (2007). Extracellular control of TGFbeta signalling in vascular development and disease. Nat. Rev. Mol. Cell Biol. 8, 857–869.
Extracellular control of TGFbeta signalling in vascular development and disease.Crossref | GoogleScholarGoogle Scholar | 17895899PubMed |

Vásquez, R., Farías, M., Vega, J. L., Martin, R. S., Vecchiola, A., Casanello, P., and Sobrevia, L. (2007). D-glucose stimulation of L-arginine transport and nitric oxide synthesis results from activation of mitogen-activated protein kinases p42/44 and Smad2 requiring functional type II TGF-beta receptors in human umbilical vein endothelium. J. Cell. Physiol. 212, 626–632.
D-glucose stimulation of L-arginine transport and nitric oxide synthesis results from activation of mitogen-activated protein kinases p42/44 and Smad2 requiring functional type II TGF-beta receptors in human umbilical vein endothelium.Crossref | GoogleScholarGoogle Scholar | 17427197PubMed |

Wang, S. E., Narasanna, A., Whitell, C. W., Wu, F. Y., Friedman, D. B., and Arteaga, C. L. (2007). Convergence of p53 and transforming growth factor beta (TGFbeta) signaling on activating expression of the tumor suppressor gene maspin in mammary epithelial cells. J. Biol. Chem. 282, 5661–5669.
Convergence of p53 and transforming growth factor beta (TGFbeta) signaling on activating expression of the tumor suppressor gene maspin in mammary epithelial cells.Crossref | GoogleScholarGoogle Scholar | 17204482PubMed |

Wang, Q., Lu, W., Yin, T., and Lu, L. (2019). Calycosin suppresses TGF-β-induced epithelial-to-mesenchymal transition and migration by upregulating BATF2 to target PAI-1 via the Wnt and PI3K/Akt signaling pathways in colorectal cancer cells. J. Exp. Clin. Cancer Res. 38, 240.
Calycosin suppresses TGF-β-induced epithelial-to-mesenchymal transition and migration by upregulating BATF2 to target PAI-1 via the Wnt and PI3K/Akt signaling pathways in colorectal cancer cells.Crossref | GoogleScholarGoogle Scholar | 31174572PubMed |

Weikkolainen, K., Keski-Oja, J., and Koli, K. (2003). Expression of latent TGF-beta binding protein LTBP-1 is hormonally regulated in normal and transformed human lung fibroblasts. Growth Factors 21, 51–60.
Expression of latent TGF-beta binding protein LTBP-1 is hormonally regulated in normal and transformed human lung fibroblasts.Crossref | GoogleScholarGoogle Scholar | 14626352PubMed |

Woad, K. J., and Robinson, R. S. (2016). Luteal angiogenesis and its control. Theriogenology 86, 221–228.
Luteal angiogenesis and its control.Crossref | GoogleScholarGoogle Scholar | 27177965PubMed |

Woad, K. J., Hunter, M. G., Mann, G. E., Laird, M., Hammond, A. J., and Robinson, R. S. (2012). Fibroblast growth factor 2 is a key determinant of vascular sprouting during bovine luteal angiogenesis. Reproduction 143, 35–43.
Fibroblast growth factor 2 is a key determinant of vascular sprouting during bovine luteal angiogenesis.Crossref | GoogleScholarGoogle Scholar | 21998077PubMed |

Wongnoppavich, A., Dukaew, N., Choonate, S., and Chairatvit, K. (2017). Upregulation of maspin expression in human cervical carcinoma cells by transforming growth factor β1 through the convergence of Smad and non-Smad signaling pathways. Oncol. Lett. 13, 3646–3652.
Upregulation of maspin expression in human cervical carcinoma cells by transforming growth factor β1 through the convergence of Smad and non-Smad signaling pathways.Crossref | GoogleScholarGoogle Scholar | 28521467PubMed |

Zhang, Y. E. (2017). Non-Smad Signaling Pathways of the TGF-β Family. Cold Spring Harb. Perspect. Biol. 9, a022129.
Non-Smad Signaling Pathways of the TGF-β Family.Crossref | GoogleScholarGoogle Scholar | 27864313PubMed |

Zhang, M., Volpert, O., Shi, Y. H., and Bouck, N. (2000). Maspin is an angiogenesis inhibitor. Nat. Med. 6, 196–199.
Maspin is an angiogenesis inhibitor.Crossref | GoogleScholarGoogle Scholar | 10655109PubMed |