Skip to main content
Top
Published in: Journal of Translational Medicine 1/2021

Open Access 01-12-2021 | Fertility | Research

Effect of the spatial–temporal specific theca cell Cyp17 overexpression on the reproductive phenotype of the novel TC17 mouse

Authors: Christian Secchi, Martina Belli, Tracy N. H. Harrison, Joseph Swift, CheMyong Ko, Antoni J. Duleba, Dwayne Stupack, R. Jeffrey Chang, Shunichi Shimasaki

Published in: Journal of Translational Medicine | Issue 1/2021

Login to get access

Abstract

Background

In the ovarian follicle, the Theca Cells (TCs) have two main functions: preserving morphological integrity and, importantly, secreting steroid androgen hormones. TCs express the essential enzyme 17α-hydroxylase/17,20-desmolase (CYP17), which permits the conversion of pregnenolone and progesterone into androgens. Dysregulation of CYP17 enzyme activity due to an intrinsic ovarian defect is hypothesized to be a cause of hyperandrogenism in women. Androgen excess is observed in women with polycystic ovary syndrome (PCOS) resulting from excess endogenous androgen production, and in transgender males undergoing exogenous testosterone therapy after female sex assignment at birth. However, the molecular and morphological effects of Cyp17 overexpression and androgen excess on folliculogenesis is unknown.

Methods

In this work, seeking a comprehensive profiling of the local outcomes of the androgen excess in the ovary, we generated a transgenic mouse model (TC17) with doxycycline (Dox)-induced Cyp17 overexpression in a local and temporal manner. TC17 mice were obtained by a combination of the Tet-dependent expression system and the Cre/LoxP gene control system.

Results

Ovaries of Dox-treated TC17 mice overexpressed Cyp17 specifically in TCs, inducing high testosterone levels. Surprisingly, TC17 ovarian morphology resembled the human ovarian features of testosterone-treated transgender men (partially impaired folliculogenesis, hypertrophic or luteinized stromal cells, atretic follicles, and collapsed clusters). We additionally assessed TC17 fertility denoting a perturbation of the normal reproductive functions (e.g., low pregnancy rate and numbers of pups per litter). Finally, RNAseq analysis permitted us to identify dysregulated genes (Lhcgr, Fshr, Runx1) and pathways (Extra Cellular Matrix and Steroid Synthesis).

Conclusions

Our novel mouse model is a versatile tool to provide innovative insights into study the effects of Cyp17 overexpression and hyperandrogenism in the ovary.
Appendix
Available only for authorised users
Literature
1.
go back to reference McNatty KP, Heath DA, Lundy T, Fidler AE, Quirke L, O’Connell A, Smith P, Groome N, Tisdall DJ. Control of early ovarian follicular development. J Reprod Fertil Suppl. 1999;54:3–16.PubMed McNatty KP, Heath DA, Lundy T, Fidler AE, Quirke L, O’Connell A, Smith P, Groome N, Tisdall DJ. Control of early ovarian follicular development. J Reprod Fertil Suppl. 1999;54:3–16.PubMed
2.
go back to reference McNatty KP, Moore LG, Hudson NL, Quirke LD, Lawrence SB, Reader K, Hanrahan JP, Smith P, Groome NP, Laitinen M, Ritvos O, Juengel JL. The oocyte and its role in regulating ovulation rate: a new paradigm in reproductive biology. Reproduction. 2004;128(4):379–86.PubMedCrossRef McNatty KP, Moore LG, Hudson NL, Quirke LD, Lawrence SB, Reader K, Hanrahan JP, Smith P, Groome NP, Laitinen M, Ritvos O, Juengel JL. The oocyte and its role in regulating ovulation rate: a new paradigm in reproductive biology. Reproduction. 2004;128(4):379–86.PubMedCrossRef
3.
go back to reference Tajima K, Orisaka M, Yata H, Goto K, Hosokawa K, Kotsuji F. Role of granulosa and theca cell interactions in ovarian follicular maturation. Microsc Res Tech. 2006;69(6):450–8.PubMedCrossRef Tajima K, Orisaka M, Yata H, Goto K, Hosokawa K, Kotsuji F. Role of granulosa and theca cell interactions in ovarian follicular maturation. Microsc Res Tech. 2006;69(6):450–8.PubMedCrossRef
4.
go back to reference Orisaka M, Tajima K, Tsang BK, Kotsuji F. Oocyte-granulosa-theca cell interactions during preantral follicular development. J Ovarian Res. 2009;2(1):9.PubMedPubMedCentralCrossRef Orisaka M, Tajima K, Tsang BK, Kotsuji F. Oocyte-granulosa-theca cell interactions during preantral follicular development. J Ovarian Res. 2009;2(1):9.PubMedPubMedCentralCrossRef
5.
go back to reference McNatty KP, Makris A, DeGrazia C, Osathanondh R, Ryan KJ. The production of progesterone, androgens, and estrogens by granulosa cells, thecal tissue, and stromal tissue from human ovaries in vitro. J Clin Endocrinol Metab. 1979;49(5):687–99.PubMedCrossRef McNatty KP, Makris A, DeGrazia C, Osathanondh R, Ryan KJ. The production of progesterone, androgens, and estrogens by granulosa cells, thecal tissue, and stromal tissue from human ovaries in vitro. J Clin Endocrinol Metab. 1979;49(5):687–99.PubMedCrossRef
6.
go back to reference Young JM, McNeilly AS. Theca: the forgotten cell of the ovarian follicle. Reproduction. 2010;140(4):489–504.PubMedCrossRef Young JM, McNeilly AS. Theca: the forgotten cell of the ovarian follicle. Reproduction. 2010;140(4):489–504.PubMedCrossRef
7.
go back to reference Richards JS, Ren YA, Candelaria N, Adams JE, Rajkovic A. Ovarian follicular theca cell recruitment, differentiation, and impact on fertility: 2017 update. Endocr Rev. 2018;39(1):1–20.PubMedCrossRef Richards JS, Ren YA, Candelaria N, Adams JE, Rajkovic A. Ovarian follicular theca cell recruitment, differentiation, and impact on fertility: 2017 update. Endocr Rev. 2018;39(1):1–20.PubMedCrossRef
8.
go back to reference Erickson GF, Magoffin DA, Dyer CA, Hofeditz C. The ovarian androgen producing cells: a review of structure/function relationships. Endocr Rev. 1985;6(3):371–99.PubMedCrossRef Erickson GF, Magoffin DA, Dyer CA, Hofeditz C. The ovarian androgen producing cells: a review of structure/function relationships. Endocr Rev. 1985;6(3):371–99.PubMedCrossRef
9.
go back to reference Ko C, Gieske MC, Al-Alem L, Hahn Y, Su W, Gong MC, Iglarz M, Koo Y. Endothelin-2 in ovarian follicle rupture. Endocrinology. 2006;147(4):1770–9.PubMedCrossRef Ko C, Gieske MC, Al-Alem L, Hahn Y, Su W, Gong MC, Iglarz M, Koo Y. Endothelin-2 in ovarian follicle rupture. Endocrinology. 2006;147(4):1770–9.PubMedCrossRef
10.
go back to reference Roberts AJ, Skinner MK. Mesenchymal-epithelial cell interactions in the ovary: estrogen-induced theca cell steroidogenesis. Mol Cell Endocrinol. 1990;72(1):R1-5.PubMedCrossRef Roberts AJ, Skinner MK. Mesenchymal-epithelial cell interactions in the ovary: estrogen-induced theca cell steroidogenesis. Mol Cell Endocrinol. 1990;72(1):R1-5.PubMedCrossRef
11.
go back to reference Rosenfield RL, Ehrmann DA. The pathogenesis of polycystic ovary syndrome (PCOS): the hypothesis of PCOS as functional ovarian hyperandrogenism revisited. Endocr Rev. 2016;37(5):467–520.PubMedPubMedCentralCrossRef Rosenfield RL, Ehrmann DA. The pathogenesis of polycystic ovary syndrome (PCOS): the hypothesis of PCOS as functional ovarian hyperandrogenism revisited. Endocr Rev. 2016;37(5):467–520.PubMedPubMedCentralCrossRef
12.
go back to reference Azziz R, Carmina E, Dewailly D, Diamanti-Kandarakis E, Escobar-Morreale HF, Futterweit W, Janssen OE, Legro RS, Norman RJ, Taylor AE, Witchel SF, Task Force on the Phenotype of the Polycystic Ovary Syndrome of The Androgen E, Society P. The Androgen excess and PCOS society criteria for the polycystic ovary syndrome: the complete task force report. Fertil Steril. 2009;91(2):456–88.PubMedCrossRef Azziz R, Carmina E, Dewailly D, Diamanti-Kandarakis E, Escobar-Morreale HF, Futterweit W, Janssen OE, Legro RS, Norman RJ, Taylor AE, Witchel SF, Task Force on the Phenotype of the Polycystic Ovary Syndrome of The Androgen E, Society P. The Androgen excess and PCOS society criteria for the polycystic ovary syndrome: the complete task force report. Fertil Steril. 2009;91(2):456–88.PubMedCrossRef
13.
go back to reference Yang MY, Fortune JE. Testosterone stimulates the primary to secondary follicle transition in bovine follicles in vitro. Biol Reprod. 2006;75(6):924–32.PubMedCrossRef Yang MY, Fortune JE. Testosterone stimulates the primary to secondary follicle transition in bovine follicles in vitro. Biol Reprod. 2006;75(6):924–32.PubMedCrossRef
14.
go back to reference Franks S. Animal models and the developmental origins of polycystic ovary syndrome: increasing evidence for the role of androgens in programming reproductive and metabolic dysfunction. Endocrinology. 2012;153(6):2536–8.PubMedCrossRef Franks S. Animal models and the developmental origins of polycystic ovary syndrome: increasing evidence for the role of androgens in programming reproductive and metabolic dysfunction. Endocrinology. 2012;153(6):2536–8.PubMedCrossRef
15.
go back to reference Caldwell AS, Middleton LJ, Jimenez M, Desai R, McMahon AC, Allan CM, Handelsman DJ, Walters KA. Characterization of reproductive, metabolic, and endocrine features of polycystic ovary syndrome in female hyperandrogenic mouse models. Endocrinology. 2014;155(8):3146–59.PubMedCrossRef Caldwell AS, Middleton LJ, Jimenez M, Desai R, McMahon AC, Allan CM, Handelsman DJ, Walters KA. Characterization of reproductive, metabolic, and endocrine features of polycystic ovary syndrome in female hyperandrogenic mouse models. Endocrinology. 2014;155(8):3146–59.PubMedCrossRef
16.
go back to reference Vendola KA, Zhou J, Adesanya OO, Weil SJ, Bondy CA. Androgens stimulate early stages of follicular growth in the primate ovary. J Clin Invest. 1998;101(12):2622–9.PubMedPubMedCentralCrossRef Vendola KA, Zhou J, Adesanya OO, Weil SJ, Bondy CA. Androgens stimulate early stages of follicular growth in the primate ovary. J Clin Invest. 1998;101(12):2622–9.PubMedPubMedCentralCrossRef
17.
go back to reference Harlow CR, Shaw HJ, Hillier SG, Hodges JK. Factors influencing follicle-stimulating hormone-responsive steroidogenesis in marmoset granulosa cells: effects of androgens and the stage of follicular maturity. Endocrinology. 1988;122(6):2780–7.PubMedCrossRef Harlow CR, Shaw HJ, Hillier SG, Hodges JK. Factors influencing follicle-stimulating hormone-responsive steroidogenesis in marmoset granulosa cells: effects of androgens and the stage of follicular maturity. Endocrinology. 1988;122(6):2780–7.PubMedCrossRef
18.
go back to reference Abbott DH, Barnett DK, Levine JE, Padmanabhan V, Dumesic DA, Jacoris S, Tarantal AF. Endocrine antecedents of polycystic ovary syndrome in fetal and infant prenatally androgenized female rhesus monkeys. Biol Reprod. 2008;79(1):154–63.PubMedCrossRef Abbott DH, Barnett DK, Levine JE, Padmanabhan V, Dumesic DA, Jacoris S, Tarantal AF. Endocrine antecedents of polycystic ovary syndrome in fetal and infant prenatally androgenized female rhesus monkeys. Biol Reprod. 2008;79(1):154–63.PubMedCrossRef
19.
go back to reference Gleicher N, Weghofer A, Barad DH. The role of androgens in follicle maturation and ovulation induction: friend or foe of infertility treatment? Reprod Biol Endocrinol. 2011;9:116.PubMedPubMedCentralCrossRef Gleicher N, Weghofer A, Barad DH. The role of androgens in follicle maturation and ovulation induction: friend or foe of infertility treatment? Reprod Biol Endocrinol. 2011;9:116.PubMedPubMedCentralCrossRef
20.
go back to reference Smith DM, Tenney DY. Effects of steroids on mouse oocyte maturation in vitro. J Reprod Fertil. 1980;60(2):331–8.PubMedCrossRef Smith DM, Tenney DY. Effects of steroids on mouse oocyte maturation in vitro. J Reprod Fertil. 1980;60(2):331–8.PubMedCrossRef
21.
go back to reference Eppig JJ, Freter RR, Ward-Bailey PF, Schultz RM. Inhibition of oocyte maturation in the mouse: participation of cAMP, steroid hormones, and a putative maturation-inhibitory factor. Dev Biol. 1983;100(1):39–49.PubMedCrossRef Eppig JJ, Freter RR, Ward-Bailey PF, Schultz RM. Inhibition of oocyte maturation in the mouse: participation of cAMP, steroid hormones, and a putative maturation-inhibitory factor. Dev Biol. 1983;100(1):39–49.PubMedCrossRef
22.
go back to reference Schultz RM, Montgomery RR, Ward-Bailey PF, Eppig JJ. Regulation of oocyte maturation in the mouse: possible roles of intercellular communication, cAMP, and testosterone. Dev Biol. 1983;95(2):294–304.PubMedCrossRef Schultz RM, Montgomery RR, Ward-Bailey PF, Eppig JJ. Regulation of oocyte maturation in the mouse: possible roles of intercellular communication, cAMP, and testosterone. Dev Biol. 1983;95(2):294–304.PubMedCrossRef
23.
go back to reference Anderiesz C, Trounson AO. The effect of testosterone on the maturation and developmental capacity of murine oocytes in vitro. Hum Reprod. 1995;10(9):2377–81.PubMedCrossRef Anderiesz C, Trounson AO. The effect of testosterone on the maturation and developmental capacity of murine oocytes in vitro. Hum Reprod. 1995;10(9):2377–81.PubMedCrossRef
24.
go back to reference Kimura S, Matsumoto T, Matsuyama R, Shiina H, Sato T, Takeyama K, Kato S. Androgen receptor function in folliculogenesis and its clinical implication in premature ovarian failure. Trends Endocrinol Metab. 2007;18(5):183–9.PubMedCrossRef Kimura S, Matsumoto T, Matsuyama R, Shiina H, Sato T, Takeyama K, Kato S. Androgen receptor function in folliculogenesis and its clinical implication in premature ovarian failure. Trends Endocrinol Metab. 2007;18(5):183–9.PubMedCrossRef
25.
go back to reference Li M, Schatten H, Sun QY. Androgen receptor’s destiny in mammalian oocytes: a new hypothesis. Mol Hum Reprod. 2009;15(3):149–54.PubMedCrossRef Li M, Schatten H, Sun QY. Androgen receptor’s destiny in mammalian oocytes: a new hypothesis. Mol Hum Reprod. 2009;15(3):149–54.PubMedCrossRef
26.
go back to reference Kidder GM, Vanderhyden BC. Bidirectional communication between oocytes and follicle cells: ensuring oocyte developmental competence. Can J Physiol Pharmacol. 2010;88(4):399–413.PubMedPubMedCentralCrossRef Kidder GM, Vanderhyden BC. Bidirectional communication between oocytes and follicle cells: ensuring oocyte developmental competence. Can J Physiol Pharmacol. 2010;88(4):399–413.PubMedPubMedCentralCrossRef
27.
go back to reference Liu X, Qiao P, Jiang A, Jiang J, Han H, Wang L, Ren C. Paracrine regulation of steroidogenesis in theca cells by granulosa cells derived from mouse preantral follicles. Biomed Res Int. 2015;2015: 925691.PubMedPubMedCentral Liu X, Qiao P, Jiang A, Jiang J, Han H, Wang L, Ren C. Paracrine regulation of steroidogenesis in theca cells by granulosa cells derived from mouse preantral follicles. Biomed Res Int. 2015;2015: 925691.PubMedPubMedCentral
28.
go back to reference Tajima K, Orisaka M, Mori T, Kotsuji F. Ovarian theca cells in follicular function. Reprod Biomed Online. 2007;15(5):591–609.PubMedCrossRef Tajima K, Orisaka M, Mori T, Kotsuji F. Ovarian theca cells in follicular function. Reprod Biomed Online. 2007;15(5):591–609.PubMedCrossRef
29.
go back to reference Tetsuka M, Hillier SG. Androgen receptor gene expression in rat granulosa cells: the role of follicle-stimulating hormone and steroid hormones. Endocrinology. 1996;137(10):4392–7.PubMedCrossRef Tetsuka M, Hillier SG. Androgen receptor gene expression in rat granulosa cells: the role of follicle-stimulating hormone and steroid hormones. Endocrinology. 1996;137(10):4392–7.PubMedCrossRef
30.
go back to reference Jakimiuk AJ, Weitsman SR, Navab A, Magoffin DA. Luteinizing hormone receptor, steroidogenesis acute regulatory protein, and steroidogenic enzyme messenger ribonucleic acids are overexpressed in thecal and granulosa cells from polycystic ovaries. J Clin Endocrinol Metab. 2001;86(3):1318–23.PubMed Jakimiuk AJ, Weitsman SR, Navab A, Magoffin DA. Luteinizing hormone receptor, steroidogenesis acute regulatory protein, and steroidogenic enzyme messenger ribonucleic acids are overexpressed in thecal and granulosa cells from polycystic ovaries. J Clin Endocrinol Metab. 2001;86(3):1318–23.PubMed
31.
go back to reference Manneras L, Cajander S, Holmang A, Seleskovic Z, Lystig T, Lonn M, Stener-Victorin E. A new rat model exhibiting both ovarian and metabolic characteristics of polycystic ovary syndrome. Endocrinology. 2007;148(8):3781–91.PubMedCrossRef Manneras L, Cajander S, Holmang A, Seleskovic Z, Lystig T, Lonn M, Stener-Victorin E. A new rat model exhibiting both ovarian and metabolic characteristics of polycystic ovary syndrome. Endocrinology. 2007;148(8):3781–91.PubMedCrossRef
32.
go back to reference Akhtar MK, Kelly SL, Kaderbhai MA. Cytochrome b(5) modulation of 17{alpha} hydroxylase and 17–20 lyase (CYP17) activities in steroidogenesis. J Endocrinol. 2005;187(2):267–74.PubMedCrossRef Akhtar MK, Kelly SL, Kaderbhai MA. Cytochrome b(5) modulation of 17{alpha} hydroxylase and 17–20 lyase (CYP17) activities in steroidogenesis. J Endocrinol. 2005;187(2):267–74.PubMedCrossRef
33.
go back to reference Patel SS, Beshay VE, Escobar JC, Carr BR. 17alpha-hydroxylase (CYP17) expression and subsequent androstenedione production in the human ovary. Reprod Sci. 2010;17(11):978–86.PubMedCrossRef Patel SS, Beshay VE, Escobar JC, Carr BR. 17alpha-hydroxylase (CYP17) expression and subsequent androstenedione production in the human ovary. Reprod Sci. 2010;17(11):978–86.PubMedCrossRef
34.
go back to reference McAllister JM, Kerin JF, Trant JM, Estabrook RW, Mason JI, Waterman MR, Simpson ER. Regulation of cholesterol side-chain cleavage and 17 alpha-hydroxylase/lyase activities in proliferating human theca interna cells in long term monolayer culture. Endocrinology. 1989;125(4):1959–66.PubMedCrossRef McAllister JM, Kerin JF, Trant JM, Estabrook RW, Mason JI, Waterman MR, Simpson ER. Regulation of cholesterol side-chain cleavage and 17 alpha-hydroxylase/lyase activities in proliferating human theca interna cells in long term monolayer culture. Endocrinology. 1989;125(4):1959–66.PubMedCrossRef
35.
go back to reference Su AI, Cooke MP, Ching KA, Hakak Y, Walker JR, Wiltshire T, Orth AP, Vega RG, Sapinoso LM, Moqrich A, Patapoutian A, Hampton GM, Schultz PG, Hogenesch JB. Large-scale analysis of the human and mouse transcriptomes. Proc Natl Acad Sci USA. 2002;99(7):4465–70.PubMedPubMedCentralCrossRef Su AI, Cooke MP, Ching KA, Hakak Y, Walker JR, Wiltshire T, Orth AP, Vega RG, Sapinoso LM, Moqrich A, Patapoutian A, Hampton GM, Schultz PG, Hogenesch JB. Large-scale analysis of the human and mouse transcriptomes. Proc Natl Acad Sci USA. 2002;99(7):4465–70.PubMedPubMedCentralCrossRef
36.
go back to reference Zhang P, Compagnone NA, Fiore C, Vigne JL, Culp P, Musci TJ, Mellon SH. Developmental gonadal expression of the transcription factor SET and its target gene, P450c17 (17alpha-hydroxylase/c17,20 lyase). DNA Cell Biol. 2001;20(10):613–24.PubMedCrossRef Zhang P, Compagnone NA, Fiore C, Vigne JL, Culp P, Musci TJ, Mellon SH. Developmental gonadal expression of the transcription factor SET and its target gene, P450c17 (17alpha-hydroxylase/c17,20 lyase). DNA Cell Biol. 2001;20(10):613–24.PubMedCrossRef
37.
go back to reference Rosenfield RL, Barnes RB, Cara JF, Lucky AW. Dysregulation of cytochrome P450c 17 alpha as the cause of polycystic ovarian syndrome. Fertil Steril. 1990;53(5):785–91.PubMedCrossRef Rosenfield RL, Barnes RB, Cara JF, Lucky AW. Dysregulation of cytochrome P450c 17 alpha as the cause of polycystic ovarian syndrome. Fertil Steril. 1990;53(5):785–91.PubMedCrossRef
38.
go back to reference Gilling-Smith C, Story H, Rogers V, Franks S. Evidence for a primary abnormality of thecal cell steroidogenesis in the polycystic ovary syndrome. Clin Endocrinol. 1997;47(1):93–9.CrossRef Gilling-Smith C, Story H, Rogers V, Franks S. Evidence for a primary abnormality of thecal cell steroidogenesis in the polycystic ovary syndrome. Clin Endocrinol. 1997;47(1):93–9.CrossRef
39.
go back to reference Nelson VL, Legro RS, Strauss JF 3rd, McAllister JM. Augmented androgen production is a stable steroidogenic phenotype of propagated theca cells from polycystic ovaries. Mol Endocrinol. 1999;13(6):946–57.PubMedCrossRef Nelson VL, Legro RS, Strauss JF 3rd, McAllister JM. Augmented androgen production is a stable steroidogenic phenotype of propagated theca cells from polycystic ovaries. Mol Endocrinol. 1999;13(6):946–57.PubMedCrossRef
40.
go back to reference Comim FV, Teerds K, Hardy K, Franks S. Increased protein expression of LHCG receptor and 17alpha-hydroxylase/17-20-lyase in human polycystic ovaries. Hum Reprod. 2013;28(11):3086–92.PubMedCrossRef Comim FV, Teerds K, Hardy K, Franks S. Increased protein expression of LHCG receptor and 17alpha-hydroxylase/17-20-lyase in human polycystic ovaries. Hum Reprod. 2013;28(11):3086–92.PubMedCrossRef
41.
go back to reference Pache TD, Chadha S, Gooren LJ, Hop WC, Jaarsma KW, Dommerholt HB, Fauser BC. Ovarian morphology in long-term androgen-treated female to male transsexuals. A human model for the study of polycystic ovarian syndrome? Histopathology. 1991;19(5):445–52.PubMedCrossRef Pache TD, Chadha S, Gooren LJ, Hop WC, Jaarsma KW, Dommerholt HB, Fauser BC. Ovarian morphology in long-term androgen-treated female to male transsexuals. A human model for the study of polycystic ovarian syndrome? Histopathology. 1991;19(5):445–52.PubMedCrossRef
42.
go back to reference Spinder T, Spijkstra JJ, Gooren LJ, Hompes PG, van Kessel H. Effects of long-term testosterone administration on gonadotropin secretion in agonadal female to male transsexuals compared with hypogonadal and normal women. J Clin Endocrinol Metab. 1989;68(1):200–7.PubMedCrossRef Spinder T, Spijkstra JJ, Gooren LJ, Hompes PG, van Kessel H. Effects of long-term testosterone administration on gonadotropin secretion in agonadal female to male transsexuals compared with hypogonadal and normal women. J Clin Endocrinol Metab. 1989;68(1):200–7.PubMedCrossRef
44.
go back to reference Kauffman AS, Thackray VG, Ryan GE, Tolson KP, Glidewell-Kenney CA, Semaan SJ, Poling MC, Iwata N, Breen KM, Duleba AJ, Stener-Victorin E, Shimasaki S, Webster NJ, Mellon PL. A novel letrozole model recapitulates both the reproductive and metabolic phenotypes of polycystic ovary syndrome in female mice. Biol Reprod. 2015;93(3):69.PubMedPubMedCentralCrossRef Kauffman AS, Thackray VG, Ryan GE, Tolson KP, Glidewell-Kenney CA, Semaan SJ, Poling MC, Iwata N, Breen KM, Duleba AJ, Stener-Victorin E, Shimasaki S, Webster NJ, Mellon PL. A novel letrozole model recapitulates both the reproductive and metabolic phenotypes of polycystic ovary syndrome in female mice. Biol Reprod. 2015;93(3):69.PubMedPubMedCentralCrossRef
46.
go back to reference Maliqueo M, Sun M, Johansson J, Benrick A, Labrie F, Svensson H, Lonn M, Duleba AJ, Stener-Victorin E. Continuous administration of a P450 aromatase inhibitor induces polycystic ovary syndrome with a metabolic and endocrine phenotype in female rats at adult age. Endocrinology. 2013;154(1):434–45.PubMedCrossRef Maliqueo M, Sun M, Johansson J, Benrick A, Labrie F, Svensson H, Lonn M, Duleba AJ, Stener-Victorin E. Continuous administration of a P450 aromatase inhibitor induces polycystic ovary syndrome with a metabolic and endocrine phenotype in female rats at adult age. Endocrinology. 2013;154(1):434–45.PubMedCrossRef
47.
go back to reference Belteki G, Haigh J, Kabacs N, Haigh K, Sison K, Costantini F, Whitsett J, Quaggin SE, Nagy A. Conditional and inducible transgene expression in mice through the combinatorial use of cre-mediated recombination and tetracycline induction. Nucleic Acids Res. 2005;33(5): e51.PubMedPubMedCentralCrossRef Belteki G, Haigh J, Kabacs N, Haigh K, Sison K, Costantini F, Whitsett J, Quaggin SE, Nagy A. Conditional and inducible transgene expression in mice through the combinatorial use of cre-mediated recombination and tetracycline induction. Nucleic Acids Res. 2005;33(5): e51.PubMedPubMedCentralCrossRef
48.
go back to reference Yu HM, Liu B, Chiu SY, Costantini F, Hsu W. Development of a unique system for spatiotemporal and lineage-specific gene expression in mice. Proc Natl Acad Sci USA. 2005;102(24):8615–20.PubMedPubMedCentralCrossRef Yu HM, Liu B, Chiu SY, Costantini F, Hsu W. Development of a unique system for spatiotemporal and lineage-specific gene expression in mice. Proc Natl Acad Sci USA. 2005;102(24):8615–20.PubMedPubMedCentralCrossRef
49.
go back to reference El Andaloussi A, Graves S, Meng F, Mandal M, Mashayekhi M, Aifantis I. Hedgehog signaling controls thymocyte progenitor homeostasis and differentiation in the thymus. Nat Immunol. 2006;7(4):418–26.PubMedCrossRef El Andaloussi A, Graves S, Meng F, Mandal M, Mashayekhi M, Aifantis I. Hedgehog signaling controls thymocyte progenitor homeostasis and differentiation in the thymus. Nat Immunol. 2006;7(4):418–26.PubMedCrossRef
50.
go back to reference Maes C, Goossens S, Bartunkova S, Drogat B, Coenegrachts L, Stockmans I, Moermans K, Nyabi O, Haigh K, Naessens M, Haenebalcke L, Tuckermann JP, Tjwa M, Carmeliet P, Mandic V, David JP, Behrens A, Nagy A, Carmeliet G, Haigh JJ. Increased skeletal VEGF enhances beta-catenin activity and results in excessively ossified bones. EMBO J. 2010;29(2):424–41.PubMedCrossRef Maes C, Goossens S, Bartunkova S, Drogat B, Coenegrachts L, Stockmans I, Moermans K, Nyabi O, Haigh K, Naessens M, Haenebalcke L, Tuckermann JP, Tjwa M, Carmeliet P, Mandic V, David JP, Behrens A, Nagy A, Carmeliet G, Haigh JJ. Increased skeletal VEGF enhances beta-catenin activity and results in excessively ossified bones. EMBO J. 2010;29(2):424–41.PubMedCrossRef
51.
go back to reference Pan W, Jin Y, Stanger B, Kiernan AE. Notch signaling is required for the generation of hair cells and supporting cells in the mammalian inner ear. Proc Natl Acad Sci USA. 2010;107(36):15798–803.PubMedPubMedCentralCrossRef Pan W, Jin Y, Stanger B, Kiernan AE. Notch signaling is required for the generation of hair cells and supporting cells in the mammalian inner ear. Proc Natl Acad Sci USA. 2010;107(36):15798–803.PubMedPubMedCentralCrossRef
52.
go back to reference Parsa S, Ramasamy SK, De Langhe S, Gupte VV, Haigh JJ, Medina D, Bellusci S. Terminal end bud maintenance in mammary gland is dependent upon FGFR2b signaling. Dev Biol. 2008;317(1):121–31.PubMedCrossRef Parsa S, Ramasamy SK, De Langhe S, Gupte VV, Haigh JJ, Medina D, Bellusci S. Terminal end bud maintenance in mammary gland is dependent upon FGFR2b signaling. Dev Biol. 2008;317(1):121–31.PubMedCrossRef
53.
go back to reference Eshkar-Oren I, Viukov SV, Salameh S, Krief S, Oh CD, Akiyama H, Gerber HP, Ferrara N, Zelzer E. The forming limb skeleton serves as a signaling center for limb vasculature patterning via regulation of Vegf. Development. 2009;136(8):1263–72.PubMedCrossRef Eshkar-Oren I, Viukov SV, Salameh S, Krief S, Oh CD, Akiyama H, Gerber HP, Ferrara N, Zelzer E. The forming limb skeleton serves as a signaling center for limb vasculature patterning via regulation of Vegf. Development. 2009;136(8):1263–72.PubMedCrossRef
54.
go back to reference Noah TK, Kazanjian A, Whitsett J, Shroyer NF. SAM pointed domain ETS factor (SPDEF) regulates terminal differentiation and maturation of intestinal goblet cells. Exp Cell Res. 2010;316(3):452–65.PubMedCrossRef Noah TK, Kazanjian A, Whitsett J, Shroyer NF. SAM pointed domain ETS factor (SPDEF) regulates terminal differentiation and maturation of intestinal goblet cells. Exp Cell Res. 2010;316(3):452–65.PubMedCrossRef
55.
go back to reference Pi M, Chen L, Huang M, Luo Q, Quarles LD. Parathyroid-specific interaction of the calcium-sensing receptor and G alpha q. Kidney Int. 2008;74(12):1548–56.PubMedPubMedCentralCrossRef Pi M, Chen L, Huang M, Luo Q, Quarles LD. Parathyroid-specific interaction of the calcium-sensing receptor and G alpha q. Kidney Int. 2008;74(12):1548–56.PubMedPubMedCentralCrossRef
56.
go back to reference Tang S, Snider P, Firulli AB, Conway SJ. Trigenic neural crest-restricted Smad7 over-expression results in congenital craniofacial and cardiovascular defects. Dev Biol. 2010;344(1):233–47.PubMedPubMedCentralCrossRef Tang S, Snider P, Firulli AB, Conway SJ. Trigenic neural crest-restricted Smad7 over-expression results in congenital craniofacial and cardiovascular defects. Dev Biol. 2010;344(1):233–47.PubMedPubMedCentralCrossRef
57.
go back to reference Wehn AK, Chapman DL. Tbx18 and Tbx15 null-like phenotypes in mouse embryos expressing Tbx6 in somitic and lateral plate mesoderm. Dev Biol. 2010;347(2):404–13.PubMedCrossRef Wehn AK, Chapman DL. Tbx18 and Tbx15 null-like phenotypes in mouse embryos expressing Tbx6 in somitic and lateral plate mesoderm. Dev Biol. 2010;347(2):404–13.PubMedCrossRef
58.
go back to reference Xu K, Nieuwenhuis E, Cohen BL, Wang W, Canty AJ, Danska JS, Coultas L, Rossant J, Wu MY, Piscione TD, Nagy A, Gossler A, Hicks GG, Hui CC, Henkelman RM, Yu LX, Sled JG, Gridley T, Egan SE. Lunatic fringe-mediated Notch signaling is required for lung alveogenesis. Am J Physiol Lung Cell Mol Physiol. 2010;298(1):L45-56.PubMedCrossRef Xu K, Nieuwenhuis E, Cohen BL, Wang W, Canty AJ, Danska JS, Coultas L, Rossant J, Wu MY, Piscione TD, Nagy A, Gossler A, Hicks GG, Hui CC, Henkelman RM, Yu LX, Sled JG, Gridley T, Egan SE. Lunatic fringe-mediated Notch signaling is required for lung alveogenesis. Am J Physiol Lung Cell Mol Physiol. 2010;298(1):L45-56.PubMedCrossRef
59.
go back to reference Parsa S, Kuremoto K, Seidel K, Tabatabai R, Mackenzie B, Yamaza T, Akiyama K, Branch J, Koh CJ, Al Alam D, Klein OD, Bellusci S. Signaling by FGFR2b controls the regenerative capacity of adult mouse incisors. Development. 2010;137(22):3743–52.PubMedPubMedCentralCrossRef Parsa S, Kuremoto K, Seidel K, Tabatabai R, Mackenzie B, Yamaza T, Akiyama K, Branch J, Koh CJ, Al Alam D, Klein OD, Bellusci S. Signaling by FGFR2b controls the regenerative capacity of adult mouse incisors. Development. 2010;137(22):3743–52.PubMedPubMedCentralCrossRef
60.
go back to reference Bridges PJ, Koo Y, Kang DW, Hudgins-Spivey S, Lan ZJ, Xu X, DeMayo F, Cooney A, Ko C. Generation of Cyp17iCre transgenic mice and their application to conditionally delete estrogen receptor alpha (Esr1) from the ovary and testis. Genesis. 2008;46(9):499–505.PubMedPubMedCentralCrossRef Bridges PJ, Koo Y, Kang DW, Hudgins-Spivey S, Lan ZJ, Xu X, DeMayo F, Cooney A, Ko C. Generation of Cyp17iCre transgenic mice and their application to conditionally delete estrogen receptor alpha (Esr1) from the ovary and testis. Genesis. 2008;46(9):499–505.PubMedPubMedCentralCrossRef
61.
go back to reference Wang L, Feng Z, Wang X, Wang X, Zhang X. DEGseq: an R package for identifying differentially expressed genes from RNA-seq data. Bioinformatics. 2010;26(1):136–8.CrossRefPubMed Wang L, Feng Z, Wang X, Wang X, Zhang X. DEGseq: an R package for identifying differentially expressed genes from RNA-seq data. Bioinformatics. 2010;26(1):136–8.CrossRefPubMed
62.
64.
go back to reference Amirikia H, Savoy-Moore RT, Sundareson AS, Moghissi KS. The effects of long-term androgen treatment on the ovary. Fertil Steril. 1986;45(2):202–8.PubMedCrossRef Amirikia H, Savoy-Moore RT, Sundareson AS, Moghissi KS. The effects of long-term androgen treatment on the ovary. Fertil Steril. 1986;45(2):202–8.PubMedCrossRef
65.
go back to reference Spinder T, Spijkstra JJ, van den Tweel JG, Burger CW, van Kessel H, Hompes PG, Gooren LJ. The effects of long term testosterone administration on pulsatile luteinizing hormone secretion and on ovarian histology in eugonadal female to male transsexual subjects. J Clin Endocrinol Metab. 1989;69(1):151–7.PubMedCrossRef Spinder T, Spijkstra JJ, van den Tweel JG, Burger CW, van Kessel H, Hompes PG, Gooren LJ. The effects of long term testosterone administration on pulsatile luteinizing hormone secretion and on ovarian histology in eugonadal female to male transsexual subjects. J Clin Endocrinol Metab. 1989;69(1):151–7.PubMedCrossRef
66.
go back to reference Grynberg M, Fanchin R, Dubost G, Colau JC, Bremont-Weil C, Frydman R, Ayoubi JM. Histology of genital tract and breast tissue after long-term testosterone administration in a female-to-male transsexual population. Reprod Biomed Online. 2010;20(4):553–8.PubMedCrossRef Grynberg M, Fanchin R, Dubost G, Colau JC, Bremont-Weil C, Frydman R, Ayoubi JM. Histology of genital tract and breast tissue after long-term testosterone administration in a female-to-male transsexual population. Reprod Biomed Online. 2010;20(4):553–8.PubMedCrossRef
67.
go back to reference Ikeda K, Baba T, Noguchi H, Nagasawa K, Endo T, Kiya T, Saito T. Excessive androgen exposure in female-to-male transsexual persons of reproductive age induces hyperplasia of the ovarian cortex and stroma but not polycystic ovary morphology. Hum Reprod. 2013;28(2):453–61.PubMedCrossRef Ikeda K, Baba T, Noguchi H, Nagasawa K, Endo T, Kiya T, Saito T. Excessive androgen exposure in female-to-male transsexual persons of reproductive age induces hyperplasia of the ovarian cortex and stroma but not polycystic ovary morphology. Hum Reprod. 2013;28(2):453–61.PubMedCrossRef
68.
go back to reference Loverro G, Resta L, Dellino M, Edoardo DN, Cascarano MA, Loverro M, Mastrolia SA. Uterine and ovarian changes during testosterone administration in young female-to-male transsexuals. Taiwan J Obstet Gynecol. 2016;55(5):686–91.PubMedCrossRef Loverro G, Resta L, Dellino M, Edoardo DN, Cascarano MA, Loverro M, Mastrolia SA. Uterine and ovarian changes during testosterone administration in young female-to-male transsexuals. Taiwan J Obstet Gynecol. 2016;55(5):686–91.PubMedCrossRef
69.
go back to reference Reeves G. Specific stroma in the cortex and medulla of the ovary. Cell types and vascular supply in relation to follicular apparatus and ovulation. Obstet Gynecol. 1971;37(6):832–44.PubMed Reeves G. Specific stroma in the cortex and medulla of the ovary. Cell types and vascular supply in relation to follicular apparatus and ovulation. Obstet Gynecol. 1971;37(6):832–44.PubMed
70.
go back to reference Moravek MB. Gender-affirming hormone therapy for transgender men. Clin Obstet Gynecol. 2018;61(4):687–704.PubMedCrossRef Moravek MB. Gender-affirming hormone therapy for transgender men. Clin Obstet Gynecol. 2018;61(4):687–704.PubMedCrossRef
71.
go back to reference Kinnear HM, Tomaszewski CE, Chang FL, Moravek MB, Xu M, Padmanabhan V, Shikanov A. The ovarian stroma as a new frontier. Reproduction. 2020;160(3):R25–39.PubMedPubMedCentralCrossRef Kinnear HM, Tomaszewski CE, Chang FL, Moravek MB, Xu M, Padmanabhan V, Shikanov A. The ovarian stroma as a new frontier. Reproduction. 2020;160(3):R25–39.PubMedPubMedCentralCrossRef
72.
go back to reference Kinnear HM, Constance ES, David A, Marsh EE, Padmanabhan V, Shikanov A, Moravek MB. A mouse model to investigate the impact of testosterone therapy on reproduction in transgender men. Hum Reprod. 2019;34(10):2009–17.PubMedPubMedCentralCrossRef Kinnear HM, Constance ES, David A, Marsh EE, Padmanabhan V, Shikanov A, Moravek MB. A mouse model to investigate the impact of testosterone therapy on reproduction in transgender men. Hum Reprod. 2019;34(10):2009–17.PubMedPubMedCentralCrossRef
73.
go back to reference Bole-Feysot C, Goffin V, Edery M, Binart N, Kelly PA. Prolactin (PRL) and its receptor: actions, signal transduction pathways and phenotypes observed in PRL receptor knockout mice. Endocr Rev. 1998;19(3):225–68.PubMedCrossRef Bole-Feysot C, Goffin V, Edery M, Binart N, Kelly PA. Prolactin (PRL) and its receptor: actions, signal transduction pathways and phenotypes observed in PRL receptor knockout mice. Endocr Rev. 1998;19(3):225–68.PubMedCrossRef
74.
go back to reference Nicol B, Grimm SA, Chalmel F, Lecluze E, Pannetier M, Pailhoux E, Dupin-De-Beyssat E, Guiguen Y, Capel B, Yao HH. RUNX1 maintains the identity of the fetal ovary through an interplay with FOXL2. Nat Commun. 2019;10(1):5116.PubMedPubMedCentralCrossRef Nicol B, Grimm SA, Chalmel F, Lecluze E, Pannetier M, Pailhoux E, Dupin-De-Beyssat E, Guiguen Y, Capel B, Yao HH. RUNX1 maintains the identity of the fetal ovary through an interplay with FOXL2. Nat Commun. 2019;10(1):5116.PubMedPubMedCentralCrossRef
75.
go back to reference Ederveen EGT, van Hunsel F, Wondergem MJ, van Puijenbroek EP. Severe secondary polycythemia in a female-to-male transgender patient while using lifelong hormonal therapy: a patient’s perspective. Drug Saf Case Rep. 2018;5(1):6.PubMedPubMedCentralCrossRef Ederveen EGT, van Hunsel F, Wondergem MJ, van Puijenbroek EP. Severe secondary polycythemia in a female-to-male transgender patient while using lifelong hormonal therapy: a patient’s perspective. Drug Saf Case Rep. 2018;5(1):6.PubMedPubMedCentralCrossRef
76.
go back to reference Madsen MC, van Dijk D, Wiepjes CM, Conemans EB, Thijs A, den Heijer M. Erythrocytosis in a large cohort of trans men using testosterone: a long-term follow-up study on prevalence, determinants, and exposure years. J Clin Endocrinol Metab. 2021;106(6):1710–7.PubMedPubMedCentralCrossRef Madsen MC, van Dijk D, Wiepjes CM, Conemans EB, Thijs A, den Heijer M. Erythrocytosis in a large cohort of trans men using testosterone: a long-term follow-up study on prevalence, determinants, and exposure years. J Clin Endocrinol Metab. 2021;106(6):1710–7.PubMedPubMedCentralCrossRef
77.
go back to reference Hembree WC, Cohen-Kettenis PT, Gooren L, Hannema SE, Meyer WJ, Murad MH, Rosenthal SM, Safer JD, Tangpricha V, T’Sjoen GG. Endocrine treatment of gender-dysphoric/gender-incongruent persons: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2017;102(11):3869–903.PubMedCrossRef Hembree WC, Cohen-Kettenis PT, Gooren L, Hannema SE, Meyer WJ, Murad MH, Rosenthal SM, Safer JD, Tangpricha V, T’Sjoen GG. Endocrine treatment of gender-dysphoric/gender-incongruent persons: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2017;102(11):3869–903.PubMedCrossRef
78.
go back to reference Coleman E, Bockting W, Botzer M, Cohen-Kettenis P, DeCuypere G, Feldman J, Fraser L, Green J, Knudson G, Meyer WJ, Monstrey S, Adler RK, Brown GR, Devor AH, Ehrbar R, Ettner R, Eyler E, Garofalo R, Karasic DH, Lev AI, Mayer G, Meyer-Bahlburg H, Hall BP, Pfaefflin F, Rachlin K, Robinson B, Schechter LS, Tangpricha V, van Trotsenburg M, Vitale A, Winter S, Whittle S, Wylie KR, Zucker K. Standards of care for the health of transsexual, transgender, and gender-nonconforming people, version 7. Int J Transgenderism. 2012;13(4):165–232.CrossRef Coleman E, Bockting W, Botzer M, Cohen-Kettenis P, DeCuypere G, Feldman J, Fraser L, Green J, Knudson G, Meyer WJ, Monstrey S, Adler RK, Brown GR, Devor AH, Ehrbar R, Ettner R, Eyler E, Garofalo R, Karasic DH, Lev AI, Mayer G, Meyer-Bahlburg H, Hall BP, Pfaefflin F, Rachlin K, Robinson B, Schechter LS, Tangpricha V, van Trotsenburg M, Vitale A, Winter S, Whittle S, Wylie KR, Zucker K. Standards of care for the health of transsexual, transgender, and gender-nonconforming people, version 7. Int J Transgenderism. 2012;13(4):165–232.CrossRef
79.
go back to reference McFarland J, Craig W, Clarke NJ, Spratt DI. Serum testosterone concentrations remain stable between injections in patients receiving subcutaneous testosterone. J Endocr Soc. 2017;1(8):1095–103.PubMedPubMedCentralCrossRef McFarland J, Craig W, Clarke NJ, Spratt DI. Serum testosterone concentrations remain stable between injections in patients receiving subcutaneous testosterone. J Endocr Soc. 2017;1(8):1095–103.PubMedPubMedCentralCrossRef
80.
go back to reference Nakamura A, Watanabe M, Sugimoto M, Sako T, Mahmood S, Kaku H, Nasu Y, Ishii K, Nagai A, Kumon H. Dose-response analysis of testosterone replacement therapy in patients with female to male gender identity disorder. Endocr J. 2013;60(3):275–81.PubMedCrossRef Nakamura A, Watanabe M, Sugimoto M, Sako T, Mahmood S, Kaku H, Nasu Y, Ishii K, Nagai A, Kumon H. Dose-response analysis of testosterone replacement therapy in patients with female to male gender identity disorder. Endocr J. 2013;60(3):275–81.PubMedCrossRef
81.
82.
go back to reference Taub RL, Ellis SA, Neal-Perry G, Magaret AS, Prager SW, Micks EA. The effect of testosterone on ovulatory function in transmasculine individuals. Am J Obstet Gynecol. 2020;223(2):229.e221-8.CrossRef Taub RL, Ellis SA, Neal-Perry G, Magaret AS, Prager SW, Micks EA. The effect of testosterone on ovulatory function in transmasculine individuals. Am J Obstet Gynecol. 2020;223(2):229.e221-8.CrossRef
83.
go back to reference Radix A, Davis AM. Endocrine treatment of gender-dysphoric/gender-incongruent persons. JAMA. 2017;318(15):1491–2.PubMedCrossRef Radix A, Davis AM. Endocrine treatment of gender-dysphoric/gender-incongruent persons. JAMA. 2017;318(15):1491–2.PubMedCrossRef
84.
go back to reference Lapinski J, Covas T, Perkins JM, Russell K, Adkins D, Coffigny MC, Hull S. Best practices in transgender health: a clinician’s guide. Prim Care. 2018;45(4):687–703.PubMedCrossRef Lapinski J, Covas T, Perkins JM, Russell K, Adkins D, Coffigny MC, Hull S. Best practices in transgender health: a clinician’s guide. Prim Care. 2018;45(4):687–703.PubMedCrossRef
85.
go back to reference Wakefield BW, Boguszewski KE, Cheney D, Taylor JF. Patterns of fertility discussions and referrals for youth at an interdisciplinary gender clinic. LGBT Health. 2019;6(8):417–21.PubMedCrossRef Wakefield BW, Boguszewski KE, Cheney D, Taylor JF. Patterns of fertility discussions and referrals for youth at an interdisciplinary gender clinic. LGBT Health. 2019;6(8):417–21.PubMedCrossRef
87.
go back to reference Rafferty J, Committee on Psychosocial Aspects Of Child And Family Health, Committee on Adolescence, Section on Lesbian, Gay, Bisexual, and Transgender Health and Wellness. Ensuring comprehensive care and support for transgender and gender-diverse children and adolescents. Pediatrics. 2018. https://doi.org/10.1542/peds.2018-2162.CrossRefPubMed Rafferty J, Committee on Psychosocial Aspects Of Child And Family Health, Committee on Adolescence, Section on Lesbian, Gay, Bisexual, and Transgender Health and Wellness. Ensuring comprehensive care and support for transgender and gender-diverse children and adolescents. Pediatrics. 2018. https://​doi.​org/​10.​1542/​peds.​2018-2162.CrossRefPubMed
Metadata
Title
Effect of the spatial–temporal specific theca cell Cyp17 overexpression on the reproductive phenotype of the novel TC17 mouse
Authors
Christian Secchi
Martina Belli
Tracy N. H. Harrison
Joseph Swift
CheMyong Ko
Antoni J. Duleba
Dwayne Stupack
R. Jeffrey Chang
Shunichi Shimasaki
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2021
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-021-03103-x

Other articles of this Issue 1/2021

Journal of Translational Medicine 1/2021 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.