252 results on '"Lydon, JP"'
Search Results
2. Altering Coregulator Concentration by Conditional Genetic Modification: Gene Dosage of REA Is Critical for Fertility and Uterine Function.
- Author
-
Park, SH, primary, Yoon, SY, additional, Zhao, Y, additional, Liao, L, additional, Liu, Z, additional, Xu, J, additional, Lydon, JP, additional, Demayo, FJ, additional, O'Malley, BW, additional, Bagchi, MK, additional, and Katzenellenbogen, BS, additional
- Published
- 2010
- Full Text
- View/download PDF
3. β-catenin mediates glandular formation and dysregulation of β-catenin induces hyperplasia formation in the murine uterus
- Author
-
Jeong, J-W, Lee, HS, Franco, HL, Broaddus, RR, Taketo, MM, Tsai, SY, Lydon, JP, and DeMayo, FJ
- Published
- 2009
4. Conditional Ablation of Progesterone Receptor Membrane Component 2 Causes Female Premature Reproductive Senescence
- Author
-
Clark, NC, primary, Pru, CA, additional, Yee, SP, additional, Lydon, JP, additional, Peluso, JJ, additional, and Pru, JK, additional
- Published
- 2016
- Full Text
- View/download PDF
5. A mouse model for endometrioid ovarian cancer arising from the distal oviduct
- Author
-
Horst, Paul, Zee, Marten, Antonissen, Claudia, Jia, YD, DeMayo, FJ, Lydon, JP, van Deurzen, Carolien, Ewing, PC, Burger, Curt, Blok, Leen, Obstetrics & Gynecology, Anesthesiology, and Pathology
- Subjects
endocrine system ,endocrine system diseases ,SDG 3 - Good Health and Well-being ,female genital diseases and pregnancy complications - Abstract
Ovarian cancer is the deadliest gynecological malignancy in Western countries. Early detection, however, is hampered by the fact that the origin of ovarian cancer remains unclear. Knowing that in a high percentage of endometrioid ovarian cancers Wnt/beta-catenin signaling is activated, and in view of the hypothesis that ovarian cancer may originate from the distal oviduct, we studied mice in which Wnt/beta-catenin signaling was activated in Mullerian duct-derived tissues. Conditional adenomatous polyposis coli (Apc) knockout mice were used to study the activation of Wnt/beta-catenin signaling in Mullerian duct-derived organs. These Pgr(Cre/+); Apc(ex15lox/lox) mice (n = 44) were sacrificed at 10, 20, 40 and 80 weeks and uterus, oviduct, ovaries and surrounding fat tissues were assessed using immunohistochemistry. Using nuclear beta-catenin staining, Wnt/beta-catenin signaling activation was confirmed in the entire epithelium of the adult Mullerian duct (fimbriae, oviduct and endometrium), but was absent in ovarian surface epithelium cells (OSEs). Besides endometrial hyperplasia, in 87.2% of mice intraepithelial lesions of the distal oviduct were found, whereas OSEs remained unaffected. In addition, 62.5% of mice developed tumors in the distal and fimbrial part of the oviduct. In the ovaries, mainly at young age, in 16.3% of mice, simple epithelial cysts were noted, which developed further into endometrioid ovarian tumors, resembling human endometrioid ovarian cancer (27.9% of mice). Next to this, locoregional growth in the utero-ovarian ligament was also shown. Here, for the first time, mutations (activation of Wnt/beta-catenin) in the distal oviduct result in precursor lesions that develop into ovarian tumors, resembling human endometrioid ovarian cancer.
- Published
- 2014
6. Alterations in Wnt-catenin and Pten signalling play distinct roles in endometrial cancer initiation and progression
- Author
-
Zee, Marten, Jia, YD, Wang, YY, Antonissen, Claudia, Graham, Patricia, Franken, Patrick, DeMayo, FJ, Lydon, JP, Burger, Curt, Fodde, Riccardo, Blok, Leen, Immunology, Anesthesiology, Pathology, Obstetrics & Gynecology, and Developmental Biology
- Subjects
SDG 3 - Good Health and Well-being - Published
- 2013
7. Embryonic Lethality in Homozygous Human Her-2 Transgenic Mice Due to Disruption of the Pds5b Gene
- Author
-
Lydon, JP, Yong, CSM, Sharkey, J, Duscio, B, Venville, B, Wei, W-Z, Jones, RF, Slaney, CY, Arnau, GM, Papenfuss, AT, Schroeder, J, Darcy, PK, Kershaw, MH, Lydon, JP, Yong, CSM, Sharkey, J, Duscio, B, Venville, B, Wei, W-Z, Jones, RF, Slaney, CY, Arnau, GM, Papenfuss, AT, Schroeder, J, Darcy, PK, and Kershaw, MH
- Abstract
The development of antigen-targeted therapeutics is dependent on the preferential expression of tumor-associated antigens (TAA) at targetable levels on the tumor. Tumor-associated antigens can be generated de novo or can arise from altered expression of normal basal proteins, such as the up-regulation of human epidermal growth factor receptor 2 (Her2/ErbB2). To properly assess the development of Her2 therapeutics in an immune tolerant model, we previously generated a transgenic mouse model in which expression of the human Her2 protein was present in both the brain and mammary tissue. This mouse model has facilitated the development of Her2 targeted therapies in a clinically relevant and suitable model. While heterozygous Her2+/- mice appear to develop in a similar manner to wild type mice (Her2-/-), it has proven difficult to generate homozygous Her2+/+ mice, potentially due to embryonic lethality. In this study, we performed whole genome sequencing to determine if the integration site of the Her2 transgene was responsible for this lethality. Indeed, we report that the Her2 transgene had integrated into the Pds5b (precocious dissociation of sisters) gene on chromosome 5, as a 162 copy concatemer. Furthermore, our findings demonstrate that Her2+/+ mice, similar to Pds5b-/- mice, are embryonic lethal and confirm the necessity for Pds5b in embryonic development. This study confirms the value of whole genome sequencing in determining the integration site of transgenes to gain insight into associated phenotypes.
- Published
- 2015
8. Progesterone drives mammary secretory differentiation via RankL-mediated induction of Elf5 in luminal progenitor cells.
- Author
-
Lee, HJ, Gallego-Ortega, D, Ledger, A, Schramek, D, Joshi, P, Szwarc, MM, Cho, C, Lydon, JP, Khokha, R, Penninger, JM, Ormandy, CJ, Lee, HJ, Gallego-Ortega, D, Ledger, A, Schramek, D, Joshi, P, Szwarc, MM, Cho, C, Lydon, JP, Khokha, R, Penninger, JM, and Ormandy, CJ
- Abstract
Progesterone-RankL paracrine signaling has been proposed as a driver of stem cell expansion in the mammary gland, and Elf5 is essential for the differentiation of mammary epithelial progenitor cells. We demonstrate that Elf5 expression is induced by progesterone and that Elf5 and progesterone cooperate to promote alveolar development. The progesterone receptor and Elf5 are expressed in a mutually exclusive pattern, and we identify RankL as the paracrine mediator of the effects of progesterone on Elf5 expression in CD61+ progenitor cells and their consequent differentiation. Blockade of RankL action prevented progesterone-induced side branching and the expansion of Elf5(+) mature luminal cells. These findings describe a mechanism by which steroid hormones can produce the expansion of steroid hormone receptor-negative mammary epithelial cells.
- Published
- 2013
9. Progesterone induction of calcitonin expression in the murine mammary gland
- Author
-
Ismail, PM, DeMayo, FJ, Amato, P, and Lydon, JP
- Abstract
Progesterone, via its nuclear receptor, is mandatory not only for the induction and specification of mammary gland ductal side-branching and lobuloalveologenesis but also for carcinogen-induced mammary tumorigenesis. Notwithstanding these recent advances, a more comprehensive molecular explanation of progesterone-induced mammary morphogenesis is contingent upon the identification and characterization of mammary molecular targets that are responsive to the progesterone signal. Toward this goal, we report that calcitonin, a 32 amino acid peptide hormone involved in calcium homeostasis, is exclusively expressed in, and secreted from, luminal epithelial cells within the mammary gland of the pregnant mouse, and, importantly, its expression is progesterone-dependent. Conversely, the calcitonin receptor is present during all stages of post-natal mammary development examined, is localized to the myoepithelial cell lineage, and is not regulated by progesterone. Because calcitonin induction spatiotemporally correlates with increases in progesterone-induced mammary gland proliferation and structural remodeling, we posit that calcitonin - through its receptor - may be involved in one or both of these progesterone-dependent processes.
- Published
- 2004
- Full Text
- View/download PDF
10. Analysis of the steroid receptor coactivator-2 (GRIP1/NCOA2)-dependent targeted metabolome in decidualizing primary human endometrial stromal cells under glycolytic stress
- Author
-
Kommagani, R, primary, Szwarc, MM, additional, Kovanci, E, additional, Gibbons, WE, additional, Putluri, N, additional, Maity, S, additional, Creighton, CJ, additional, Sreekumar, A, additional, DeMayo, FJ, additional, Lydon, JP, additional, and O'Malley, BW, additional
- Full Text
- View/download PDF
11. A Mouse Model for Conditional Expression of Activated β-Catenin in Epidermal Keratinocytes.
- Author
-
Maurya VK, Ying Y, and Lydon JP
- Subjects
- Animals, Mice, Mice, Transgenic, Cell Proliferation, Hair Follicle metabolism, Hair Follicle growth & development, Keratin-5 metabolism, Keratin-5 genetics, Female, beta Catenin metabolism, beta Catenin genetics, Keratinocytes metabolism, Doxycycline pharmacology, Epidermis metabolism, Epidermis growth & development
- Abstract
We report the generation and characterization of the K5: CAT bigenic mouse in which the constitutively activated form of β-catenin (ΔN89 β-catenin) is conditionally expressed in cytokeratin-5 (K5) positive epidermal keratinocytes. Following short-term doxycycline intake during the telogen resting phase, the adult K5: CAT bigenic develops enlarged pilosebaceous units that expand deep into the dermis, an expansion usually observed during the anagen growth phase. Prolonged doxycycline treatment results in significant thickening and folding of the K5: CAT epidermis. During this persistent induction period, there is clear evidence of increased keratinocyte proliferation, particularly in the epidermal basal cell layer and the outer root sheath of the hair follicle. This unscheduled increase in cellular proliferation likely explains the decrease in hair density observed in the K5: CAT mouse following persistent doxycycline intake. Numerous hyperplastic endometrioid cysts, which display cornification toward their lumens, are also observed during this treatment period. Remarkably, de-induction of ΔN89 β-catenin expression through doxycycline withdrawal results in a marked reversal of the skin phenotype, suggesting that these morphological changes are dependent on continued signaling by β-catenin and/or its downstream molecular mediators. Joining a small group of mouse models for conditional β-catenin signaling, our K5: CAT mouse model will be particularly useful in identifying those molecular mediators of β-catenin that are responsible for initiating and maintaining these phenotypic responses in the K5: CAT skin. Such studies are predicted to shed more light on β-catenin signaling in epidermal epithelial morphogenesis, hair follicle cycling, and hair growth pathologies., Competing Interests: Declarations. Conflict of interests: The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Contributions of white adipose tissue to energy requirements for female reproduction.
- Author
-
Anaya ES, de Groot EL, Lydon JP, Pangas SA, and Hartig SM
- Subjects
- Humans, Female, Animals, Ovary physiology, Ovary metabolism, Adipose Tissue, White metabolism, Adipose Tissue, White physiology, Energy Metabolism physiology, Reproduction physiology
- Abstract
Body composition impacts female fertility and there are established relationships between adipose tissue and the reproductive system. Maintaining functional adipose tissue is vital for meeting the energetic demands during the reproductive process, from ovulation to delivery and lactation. White adipose tissue (WAT) shows plastic responses to daily physiology and secretes diverse adipokines that affect the hypothalamic-pituitary-ovarian axis, but many other interorgan interactions remain to be determined. This review summarizes the current state of research on the dialogue between WAT and the female reproductive system, focusing on the impact of this crosstalk on ovarian and endometrial factors essential for fecundity., Competing Interests: Declaration of interests No interests are declared., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Generation of Oviductal Glycoprotein 1 Cre Mouse Model for the Study of Secretory Epithelial Cells of the Oviduct.
- Author
-
McGlade EA, Mao J, Stephens KK, Kelleher AM, Maddison LA, Bernhardt ML, DeMayo FJ, Lydon JP, and Winuthayanon W
- Subjects
- Animals, Female, Mice, Male, Oviducts metabolism, Oviducts cytology, Mice, Transgenic, Green Fluorescent Proteins metabolism, Green Fluorescent Proteins genetics, Fallopian Tubes metabolism, Fallopian Tubes cytology, Receptors, Progesterone metabolism, Receptors, Progesterone genetics, Models, Animal, Epithelial Cells metabolism, Integrases metabolism, Integrases genetics, Glycoproteins genetics, Glycoproteins metabolism
- Abstract
The epithelial cell lining of the oviduct plays an important role in oocyte pickup, sperm migration, preimplantation embryo development, and embryo transport. The oviduct epithelial cell layer comprises ciliated and nonciliated secretory cells. The ciliary function has been shown to support gamete and embryo movement in the oviduct, yet secretory cell function has not been well characterized. Therefore, our goal was to generate a secretory cell-specific Cre recombinase mouse model to study the role of the oviductal secretory cells. A knock-in mouse model, Ovgp1Cre:eGFP, was created by expressing Cre from the endogenous Ovgp1 (oviductal glycoprotein 1) locus, with enhanced green fluorescent protein (eGFP) as a reporter. EGFP signals were strongly detected in the secretory epithelial cells of the oviducts at estrus in adult Ovgp1Cre:eGFP mice. Signals were also detected in the ovarian stroma, uterine stroma, vaginal epithelial cells, epididymal epithelial cells, and elongated spermatids. To validate recombinase activity, progesterone receptor (PGR) expression was ablated using the Ovgp1Cre:eGFP; Pgrf/f mouse model. Surprisingly, the deletion was restricted to the epithelial cells of the uterotubal junction (UTJ) region of Ovgp1Cre:eGFP; Pgrf/f oviducts. Deletion of Pgr in the epithelial cells of the UTJ region had no effect on female fecundity. In summary, we found that eGFP signals were likely specific to secretory epithelial cells in all regions of the oviduct. However, due to a potential target-specific Cre activity, validation of appropriate recombination and expression of the gene(s) of interest is absolutely required to confirm efficient deletion when generating conditional knockout mice using the Ovgp1Cre:eGFP line., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com. See the journal About page for additional terms.)
- Published
- 2024
- Full Text
- View/download PDF
14. Establishment of Murine Pregnancy Requires the Promyelocytic Leukemia Zinc Finger Transcription Factor.
- Author
-
Hai L, Maurya VK, DeMayo FJ, and Lydon JP
- Subjects
- Pregnancy, Female, Mice, Animals, Humans, Decidua metabolism, Endometrium metabolism, Mice, Knockout, Zinc Fingers, Stromal Cells metabolism, Transcription Factors metabolism, Leukemia metabolism
- Abstract
Using an established human primary cell culture model, we previously demonstrated that the promyelocytic leukemia zinc finger (PLZF) transcription factor is a direct target of the progesterone receptor (PGR) and is essential for progestin-dependent decidualization of human endometrial stromal cells (HESCs). These in vitro findings were supported by immunohistochemical analysis of human endometrial tissue biopsies, which showed that the strongest immunoreactivity for endometrial PLZF is detected during the progesterone (P4)-dominant secretory phase of the menstrual cycle. While these human studies provided critical clinical support for the important role of PLZF in P4-dependent HESC decidualization, functional validation in vivo was not possible due to the absence of suitable animal models. To address this deficiency, we recently generated a conditional knockout mouse model in which PLZF is ablated in PGR-positive cells of the mouse ( Plzf
d/d ). The Plzfd/d female was phenotypically analyzed using immunoblotting, real-time PCR, and immunohistochemistry. Reproductive function was tested using the timed natural pregnancy model as well as the artificial decidual response assay. Even though ovarian activity is not affected, female Plzfd/d mice exhibit an infertility phenotype due to an inability of the embryo to implant into the Plzfd/d endometrium. Initial cellular and molecular phenotyping investigations reveal that the Plzfd/d endometrium is unable to develop a transient receptive state, which is reflected at the molecular level by a blunted response to P4 exposure with a concomitant unopposed response to 17-β estradiol. In addition to a defect in P4-dependent receptivity, the Plzfd/d endometrium fails to undergo decidualization in response to an artificial decidual stimulus, providing the in vivo validation for our earlier HESC culture findings. Collectively, our new Plzfd/d mouse model underscores the physiological importance of the PLZF transcription factor not only in endometrial stromal cell decidualization but also uterine receptivity, two uterine cellular processes that are indispensable for the establishment of pregnancy.- Published
- 2024
- Full Text
- View/download PDF
15. A GREB1-steroid receptor feedforward mechanism governs differential GREB1 action in endometrial function and endometriosis.
- Author
-
Chadchan SB, Popli P, Liao Z, Andreas E, Dias M, Wang T, Gunderson SJ, Jimenez PT, Lanza DG, Lanz RB, Foulds CE, Monsivais D, DeMayo FJ, Yalamanchili HK, Jungheim ES, Heaney JD, Lydon JP, Moley KH, O'Malley BW, and Kommagani R
- Subjects
- Animals, Female, Humans, Mice, Endometrium metabolism, Estrogens metabolism, Progesterone metabolism, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, Steroids metabolism, Endometriosis genetics, Endometriosis metabolism, Neoplasm Proteins metabolism, Receptors, Steroid genetics, Receptors, Steroid metabolism
- Abstract
Cellular responses to the steroid hormones, estrogen (E2), and progesterone (P4) are governed by their cognate receptor's transcriptional output. However, the feed-forward mechanisms that shape cell-type-specific transcriptional fulcrums for steroid receptors are unidentified. Herein, we found that a common feed-forward mechanism between GREB1 and steroid receptors regulates the differential effect of GREB1 on steroid hormones in a physiological or pathological context. In physiological (receptive) endometrium, GREB1 controls P4-responses in uterine stroma, affecting endometrial receptivity and decidualization, while not affecting E2-mediated epithelial proliferation. Of mechanism, progesterone-induced GREB1 physically interacts with the progesterone receptor, acting as a cofactor in a positive feedback mechanism to regulate P4-responsive genes. Conversely, in endometrial pathology (endometriosis), E2-induced GREB1 modulates E2-dependent gene expression to promote the growth of endometriotic lesions in mice. This differential action of GREB1 exerted by a common feed-forward mechanism with steroid receptors advances our understanding of mechanisms that underlie cell- and tissue-specific steroid hormone actions., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
16. Steroid receptor coactivator-2 drives epithelial reprogramming that enables murine embryo implantation.
- Author
-
Maurya VK, Szwarc MM, Lonard DM, Kommagani R, Wu SP, O'Malley BW, DeMayo FJ, and Lydon JP
- Subjects
- Animals, Female, Humans, Mice, Pregnancy, Endometrium metabolism, Epithelial Cells metabolism, Epithelial-Mesenchymal Transition, Mice, Knockout, Nuclear Receptor Coactivator 2 genetics, Embryo Implantation genetics, Uterus metabolism
- Abstract
Although we have shown that steroid receptor coactivator-2 (SRC-2), a member of the p160/SRC family of transcriptional coregulators, is essential for decidualization of both human and murine endometrial stromal cells, SRC-2's role in the earlier stages of the implantation process have not been adequately addressed. Using a conditional SRC-2 knockout mouse (SRC-2
d/d ) in timed natural pregnancy studies, we show that endometrial SRC-2 is required for embryo attachment and adherence to the luminal epithelium. Implantation failure is associated with the persistent expression of Mucin 1 and E-cadherin on the apical surface and basolateral adherens junctions of the SRC-2d/d luminal epithelium, respectively. These findings indicate that the SRC-2d/d luminal epithelium fails to exhibit a plasma membrane transformation (PMT) state known to be required for the development of uterine receptivity. Transcriptomics demonstrated that the expression of genes involved in steroid hormone control of uterine receptivity were significantly disrupted in the SRC-2d/d endometrium as well as genes that control epithelial tight junctional biology and the emergence of the epithelial mesenchymal transition state, with the latter sharing similar biological properties with PMT. Collectively, these findings uncover a new role for endometrial SRC-2 in the induction of the luminal epithelial PMT state, which is a prerequisite for the development of uterine receptivity and early pregnancy establishment., (© 2023 The Authors. The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)- Published
- 2023
- Full Text
- View/download PDF
17. Functional analysis reveals driver cooperativity and novel mechanisms in endometrial carcinogenesis.
- Author
-
Brown M, Leon A, Kedzierska K, Moore C, Belnoue-Davis HL, Flach S, Lydon JP, DeMayo FJ, Lewis A, Bosse T, Tomlinson I, and Church DN
- Subjects
- Female, Humans, Mice, Animals, F-Box-WD Repeat-Containing Protein 7 genetics, F-Box-WD Repeat-Containing Protein 7 metabolism, Mutation, Mutation, Missense, Carcinogenesis genetics, Endometrial Neoplasms genetics, Endometrial Neoplasms metabolism
- Abstract
High-risk endometrial cancer has poor prognosis and is increasing in incidence. However, understanding of the molecular mechanisms which drive this disease is limited. We used genetically engineered mouse models (GEMM) to determine the functional consequences of missense and loss of function mutations in Fbxw7, Pten and Tp53, which collectively occur in nearly 90% of high-risk endometrial cancers. We show that Trp53 deletion and missense mutation cause different phenotypes, with the latter a substantially stronger driver of endometrial carcinogenesis. We also show that Fbxw7 missense mutation does not cause endometrial neoplasia on its own, but potently accelerates carcinogenesis caused by Pten loss or Trp53 missense mutation. By transcriptomic analysis, we identify LEF1 signalling as upregulated in Fbxw7/FBXW7-mutant mouse and human endometrial cancers, and in human isogenic cell lines carrying FBXW7 mutation, and validate LEF1 and the additional Wnt pathway effector TCF7L2 as novel FBXW7 substrates. Our study provides new insights into the biology of high-risk endometrial cancer and suggests that targeting LEF1 may be worthy of investigation in this treatment-resistant cancer subgroup., (© 2023 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
18. The NR2F2-HAND2 signaling axis regulates progesterone actions in the uterus at early pregnancy.
- Author
-
Oh Y, Quiroz E, Wang T, Medina-Laver Y, Redecke SM, Dominguez F, Lydon JP, DeMayo FJ, and Wu SP
- Subjects
- Female, Humans, Pregnancy, Animals, Mice, Signal Transduction, Endometrium, Orphan Nuclear Receptors, COUP Transcription Factor II, Progesterone pharmacology, Uterus
- Abstract
Endometrial function is dependent on a tight crosstalk between the epithelial and stromal cells of the endometrium. This communication is critical to ensure a fertile uterus and relies on progesterone and estrogen signaling to prepare a receptive uterus for embryo implantation in early pregnancy. One of the key mediators of this crosstalk is the orphan nuclear receptor NR2F2, which regulates uterine epithelial receptivity and stromal cell differentiation. In order to determine the molecular mechanism regulated by NR2F2, RNAseq analysis was conducted on the uterus of Pgr
Cre ;Nr2f2f/f mice at Day 3.5 of pregnancy. This transcriptomic analysis demonstrated Nr2f2 ablation in Pgr -expressing cells leads to a reduction of Hand2 expression, increased levels of Hand2 downstream effectors Fgf1 and Fgf18 , and a transcriptome manifesting suppressed progesterone signaling with an altered immune baseline. ChIPseq analysis conducted on the Day 3.5 pregnant mouse uterus for NR2F2 demonstrated the majority of NR2F2 occupies genomic regions that have H3K27ac and H3K4me1 histone modifications, including the loci of major uterine transcription regulators Hand2 , Egr1 , and Zbtb16 . Furthermore, functional analysis of an NR2F2 occupying site that is conserved between human and mouse was capable to enhance endogenous HAND2 mRNA expression with the CRISPR activator in human endometrial stroma cells. These data establish the NR2F2 dependent regulation of Hand2 in the stroma and identify a cis-acting element for this action. In summary, our findings reveal a role of the NR2F2-HAND2 regulatory axis that determines the uterine transcriptomic pattern in preparation for the endometrial receptivity., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Oh, Quiroz, Wang, Medina-Laver, Redecke, Dominguez, Lydon, DeMayo and Wu.)- Published
- 2023
- Full Text
- View/download PDF
19. Chromatin architectural factor CTCF is essential for progesterone-dependent uterine maturation.
- Author
-
Hewitt SC, Gruzdev A, Willson CJ, Wu SP, Lydon JP, Galjart N, and DeMayo FJ
- Subjects
- Humans, Female, Animals, Mice, Progesterone, Uterus, Endometrium, Chromatin, Endometrial Neoplasms
- Abstract
Receptors for estrogen and progesterone frequently interact, via Cohesin/CTCF loop extrusion, at enhancers distal from regulated genes. Loss-of-function CTCF mutation in >20% of human endometrial tumors indicates its importance in uterine homeostasis. To better understand how CTCF-mediated enhancer-gene interactions impact endometrial development and function, the Ctcf gene was selectively deleted in female reproductive tissues of mice. Prepubertal Ctcf
d/d uterine tissue exhibited a marked reduction in the number of uterine glands compared to those without Ctcf deletion (Ctcff/f mice). Post-pubertal Ctcfd/d uteri were hypoplastic with significant reduction in both the amount of the endometrial stroma and number of glands. Transcriptional profiling revealed increased expression of stem cell molecules Lif, EOMES, and Lgr5, and enhanced inflammation pathways following Ctcf deletion. Analysis of the response of the uterus to steroid hormone stimulation showed that CTCF deletion affects a subset of progesterone-responsive genes. This finding indicates (1) Progesterone-mediated signaling remains functional following Ctcf deletion and (2) certain progesterone-regulated genes are sensitive to Ctcf deletion, suggesting they depend on gene-enhancer interactions that require CTCF. The progesterone-responsive genes altered by CTCF ablation included Ihh, Fst, and Errfi1. CTCF-dependent progesterone-responsive uterine genes enhance critical processes including anti-tumorigenesis, which is relevant to the known effectiveness of progesterone in inhibiting progression of early-stage endometrial tumors. Overall, our findings reveal that uterine Ctcf plays a key role in progesterone-dependent expression of uterine genes underlying optimal post-pubertal uterine development., (© 2023 Federation of American Societies for Experimental Biology. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.)- Published
- 2023
- Full Text
- View/download PDF
20. TRIM28 modulates nuclear receptor signaling to regulate uterine function.
- Author
-
Li R, Wang T, Marquardt RM, Lydon JP, Wu SP, and DeMayo FJ
- Subjects
- Pregnancy, Female, Humans, Uterus metabolism, Progesterone metabolism, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, Epithelium metabolism, Tripartite Motif-Containing Protein 28 genetics, Tripartite Motif-Containing Protein 28 metabolism, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Estradiol metabolism
- Abstract
Estrogen and progesterone, acting through their cognate receptors the estrogen receptor α (ERα) and the progesterone receptor (PR) respectively, regulate uterine biology. Using rapid immunoprecipitation and mass spectrometry (RIME) and co-immunoprecipitation, we identified TRIM28 (Tripartite motif containing 28) as a protein which complexes with ERα and PR in the regulation of uterine function. Impairment of TRIM28 expression results in the inability of the uterus to support early pregnancy through altered PR and ERα action in the uterine epithelium and stroma by suppressing PR and ERα chromatin binding. Furthermore, TRIM28 ablation in PR-expressing uterine cells results in the enrichment of a subset of TRIM28 positive and PR negative pericytes and epithelial cells with progenitor potential. In summary, our study reveals the important roles of TRIM28 in regulating endometrial cell composition and function in women, and also implies its critical functions in other hormone regulated systems., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2023
- Full Text
- View/download PDF
21. A CRISPR/Cas9-engineered mouse carrying a conditional knockout allele for the early growth response-1 transcription factor.
- Author
-
Maurya VK, Ying Y, Lanza DG, Heaney JD, and Lydon JP
- Subjects
- Mice, Female, Animals, Alleles, Exons, Transcription Factors genetics, CRISPR-Cas Systems
- Abstract
Early growth response 1 (EGR1) mediates transcriptional programs that are indispensable for cell division, differentiation, and apoptosis in numerous physiologies and pathophysiologies. Whole-body EGR1 knockouts in mice (Egr1
KO ) have advanced our understanding of EGR1 function in an in vivo context. To extend the utility of the mouse to investigate EGR1 responses in a tissue- and/or cell-type-specific manner, we generated a mouse model in which exon 2 of the mouse Egr1 gene is floxed by CRISPR/Cas9 engineering. The floxed Egr1 alleles (Egr1f/f ) are designed to enable spatiotemporal control of Cre-mediated EGR1 ablation in the mouse. To confirm that the Egr1f/f alleles can be abrogated using a Cre driver, we crossed the Egr1f/f mouse with a global Cre driver to generate the Egr1 conditional knockout (Egr1d/d ) mouse in which EGR1 expression is ablated in all tissues. Genetic and protein analysis confirmed the absence of exon 2 and loss of EGR1 expression in the Egr1d/d mouse, respectively. Moreover, the Egr1d/d female exhibits overt reproductive phenotypes previously reported for the Egr1KO mouse. Therefore, studies described in this short technical report underscore the potential utility of the murine Egr1 floxed allele to further resolve EGR1 function at a tissue- and/or cell-type-specific level., (© 2023 Wiley Periodicals LLC.)- Published
- 2023
- Full Text
- View/download PDF
22. CFP1 governs uterine epigenetic landscapes to intervene in progesterone responses for uterine physiology and suppression of endometriosis.
- Author
-
Yang SC, Park M, Hong KH, La H, Park C, Wang P, Li G, Chen Q, Choi Y, DeMayo FJ, Lydon JP, Skalnik DG, Lim HJ, Hong SH, Park SH, Kim YS, Kim HR, and Song H
- Subjects
- Animals, Female, Humans, Mice, Pregnancy, Embryo Implantation genetics, Endometrium metabolism, Epigenesis, Genetic, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, RNA, Messenger metabolism, Uterus metabolism, Endometriosis genetics, Endometriosis metabolism, Progesterone pharmacology, Progesterone metabolism, Trans-Activators genetics
- Abstract
Progesterone (P
4 ) is required for the preparation of the endometrium for a successful pregnancy. P4 resistance is a leading cause of the pathogenesis of endometrial disorders like endometriosis, often leading to infertility; however, the underlying epigenetic cause remains unclear. Here we demonstrate that CFP1, a regulator of H3K4me3, is required for maintaining epigenetic landscapes of P4 -progesterone receptor (PGR) signaling networks in the mouse uterus. Cfp1f/f ;Pgr-Cre (Cfp1d/d ) mice showed impaired P4 responses, leading to complete failure of embryo implantation. mRNA and chromatin immunoprecipitation sequencing analyses showed that CFP1 regulates uterine mRNA profiles not only in H3K4me3-dependent but also in H3K4me3-independent manners. CFP1 directly regulates important P4 response genes, including Gata2, Sox17, and Ihh, which activate smoothened signaling pathway in the uterus. In a mouse model of endometriosis, Cfp1d/d ectopic lesions showed P4 resistance, which was rescued by a smoothened agonist. In human endometriosis, CFP1 was significantly downregulated, and expression levels between CFP1 and these P4 targets are positively related regardless of PGR levels. In brief, our study provides that CFP1 intervenes in the P4 -epigenome-transcriptome networks for uterine receptivity for embryo implantation and the pathogenesis of endometriosis., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
23. Beclin-1-dependent autophagy, but not apoptosis, is critical for stem-cell-mediated endometrial programming and the establishment of pregnancy.
- Author
-
Popli P, Tang S, Chadchan SB, Talwar C, Rucker EB 3rd, Guan X, Monsivais D, Lydon JP, Stallings CL, Moley KH, and Kommagani R
- Subjects
- Animals, Female, Humans, Mice, Pregnancy, Autophagy, Beclin-1, Stem Cells, Endometrium, Uterus
- Abstract
The human endometrium shows a remarkable regenerative capacity that enables cyclical regeneration and remodeling throughout a woman's reproductive life. Although early postnatal uterine developmental cues direct this regeneration, the vital factors that govern early endometrial programming are largely unknown. We report that Beclin-1, an essential autophagy-associated protein, plays an integral role in uterine morphogenesis during the early postnatal period. We show that conditional depletion of Beclin-1 in the uterus triggers apoptosis and causes progressive loss of Lgr5
+ /Aldh1a1+ endometrial progenitor stem cells, with concomitant loss of Wnt signaling, which is crucial for stem cell renewal and epithelial gland development. Beclin-1 knockin (Becn1 KI) mice with disabled apoptosis exhibit normal uterine development. Importantly, the restoration of Beclin-1-driven autophagy, but not apoptosis, promotes normal uterine adenogenesis and morphogenesis. Together, the data suggest that Beclin-1-mediated autophagy acts as a molecular switch that governs the early uterine morphogenetic program by maintaining the endometrial progenitor stem cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
24. Visualization of preimplantation uterine fluid absorption in mice using Alexa Fluor™ 488 Hydrazide†.
- Author
-
Li Y, Martin TE, Hancock JM, Li R, Viswanathan S, Lydon JP, Zheng Y, and Ye X
- Subjects
- Pregnancy, Female, Animals, Mice, Progesterone pharmacology, Estrogens pharmacology, Uterus physiology, Rodentia, Mifepristone pharmacology, Embryo Implantation physiology
- Abstract
Uterine fluid plays important roles in supporting early pregnancy events and its timely absorption is critical for embryo implantation. In mice, its volume is maximum on day 0.5 post-coitum (D0.5) and approaches minimum upon embryo attachment ~D4.0. Its secretion and absorption in ovariectomized rodents were shown to be promoted by estrogen and progesterone (P4), respectively. The temporal mechanisms in preimplantation uterine fluid absorption remain to be elucidated. We have established an approach using intraluminally injected Alexa Fluor™ 488 Hydrazide (AH) in preimplantation control (RhoAf/f) and P4-deficient RhoAf/fPgrCre/+ mice. In control mice, bulk entry (seen as smeared cellular staining) via uterine luminal epithelium (LE) decreases from D0.5 to D3.5. In P4-deficient RhoAf/fPgrCre/+ mice, bulk entry on D0.5 and D3.5 is impaired. Exogenous P4 treatment on D1.5 and D2.5 increases bulk entry in D3.5 P4-deficient RhoAf/fPgrCre/+ LE, while progesterone receptor (PR) antagonist RU486 treatment on D1.5 and D2.5 diminishes bulk entry in D3.5 control LE. The abundance of autofluorescent apical fine dots, presumptively endocytic vesicles to reflect endocytosis, in the LE cells is generally increased from D0.5 to D3.5 but its regulation by exogenous P4 or RU486 is not obvious under our experimental setting. In the glandular epithelium (GE), bulk entry is rarely observed and green cellular dots do not show any consistent differences among all the investigated conditions. This study demonstrates the dominant role of LE but not GE, the temporal mechanisms of bulk entry and endocytosis in the LE, and the inhibitory effects of P4-deficiency and RU486 on bulk entry in the LE in preimplantation uterine fluid absorption., (© The Author(s) 2022. Published by Oxford University Press on behalf of Society for the Study of Reproduction. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
25. Versican provides the provisional matrix for uterine spiral artery dilation and fetal growth.
- Author
-
Sagae Y, Horie A, Yanai A, Ohara T, Nakakita B, Kitawaki Y, Okunomiya A, Tani H, Yamaguchi K, Hamanishi J, Lydon JP, Daikoku T, Watanabe H, and Mandai M
- Subjects
- Pregnancy, Humans, Female, Mice, Animals, Dilatation, Fetal Development, Arteries metabolism, Mammals metabolism, Versicans genetics, Versicans metabolism, Uterus metabolism
- Abstract
The extracellular matrix (ECM) in the endometrium plays a crucial role in mammalian pregnancy. We have shown that versican secreted from the endometrial epithelium promotes embryo implantation. Versican is a proteoglycan, a major player in the provisional matrix, and versikine, its N-terminal fragment cleaved by ADAMTS proteinases, serves as a bioactive molecule. Here, since versican expression in the placenta was dynamically altered in humans and mice, we investigated the role of versican in pregnancy using uterine-specific Vcan deletion mice (uKO mice) and ADAMTS-resistant versican expressing mice (V1R mice). uKO mice exhibited insufficient spiral artery dilation, followed by fetal growth restriction and maternal hypertension. Further analysis revealed impaired proliferation of tissue-resident natural killer cells required for spiral artery dilation. V1R mice showed the same results as the control, eliminating the involvement of versikine. Our results provide a new concept that versican, one factor of ECM, contributes to placentation and following fetal growth., Competing Interests: Competing Interests statement The authors declare no conflicts of interest associated with this manuscript., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Histopathologic and transcriptomic phenotypes of a conditional RANKL transgenic mouse thymus.
- Author
-
Szwarc MM, Hai L, Maurya VK, Rajapakshe K, Perera D, Ittmann MM, Mo Q, Lin Y, Bettini ML, Coarfa C, and Lydon JP
- Subjects
- Agglutinins metabolism, Animals, Cytokines metabolism, Epithelial Cells metabolism, Forkhead Transcription Factors metabolism, Keratin-5 genetics, Keratin-5 metabolism, Ligands, Mice, Mice, Transgenic, NF-kappa B metabolism, Phenotype, Receptor Activator of Nuclear Factor-kappa B genetics, Receptor Activator of Nuclear Factor-kappa B metabolism, Thymus Gland metabolism, Doxycycline pharmacology, RANK Ligand metabolism, Transcriptome
- Abstract
Although conventional knockout and transgenic mouse models have significantly advanced our understanding of Receptor Activator of NF-κB Ligand (RANKL) signaling in intra-thymic crosstalk that establishes self-tolerance and later stages of lymphopoiesis, the unique advantages of conditional mouse transgenesis have yet to be explored. A main advantage of conditional transgenesis is the ability to express a transgene in a spatiotemporal restricted manner, enabling the induction (or de-induction) of transgene expression during predetermined stages of embryogenesis or during defined postnatal developmental or physiological states, such as puberty, adulthood, and pregnancy. Here, we describe the K5: RANKL bigenic mouse, in which transgene derived RANKL expression is induced by doxycycline and targeted to cytokeratin 5 positive medullary thymic epithelial cells (mTECs). Short-term doxycycline induction reveals that RANKL transgene expression is significantly induced in the thymic medulla and only in response to doxycycline. Prolonged doxycycline induction in the K5: RANKL bigenic results in a significantly enlarged thymus in which mTECs are hyperproliferative. Flow cytometry showed that there is a marked enrichment of CD4+ and CD8+ single positive thymocytes with a concomitant depletion of CD4+ CD8+ double positives. Furthermore, there is an increase in the number of FOXP3+ T regulatory (Treg) cells and Ulex Europaeus Agglutinin 1+ (UEA1+) mTECs. Transcriptomics revealed that a remarkable array of signals-cytokines, chemokines, growth factors, transcription factors, and morphogens-are governed by RANKL and drive in part the K5: RANKL thymic phenotype. Extended doxycycline administration to 6-weeks results in a K5: RANKL thymus that begins to display distinct histopathological features, such as medullary epithelial hyperplasia, extensive immune cell infiltration, and central tissue necrosis. As there are intense efforts to develop clinical approaches to restore thymic medullary function in the adult to treat immunopathological conditions in which immune cell function is compromised following cancer therapy or toxin exposure, an improved molecular understanding of RANKL's involvement in thymic medulla enlargement will be required. We believe the versatility of the conditional K5: RANKL mouse represents a tractable model system to assist in addressing this requirement as well as many other questions related to RANKL's role in thymic normal physiology and disease processes., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
27. Decidualization of human endometrial stromal cells requires steroid receptor coactivator-3.
- Author
-
Maurya VK, Szwarc MM, Lonard DM, Gibbons WE, Wu SP, O'Malley BW, DeMayo FJ, and Lydon JP
- Abstract
Steroid receptor coactivator-3 (SRC-3; also known as NCOA3 or AIB1) is a member of the multifunctional p160/SRC family of coactivators, which also includes SRC-1 and SRC-2. Clinical and cell-based studies as well as investigations on mice have demonstrated pivotal roles for each SRC in numerous physiological and pathophysiological contexts, underscoring their functional pleiotropy. We previously demonstrated the critical involvement of SRC-2 in murine embryo implantation as well as in human endometrial stromal cell (HESC) decidualization, a cellular transformation process required for trophoblast invasion and ultimately placentation. We show here that, like SRC-2, SRC-3 is expressed in the epithelial and stromal cellular compartments of the human endometrium during the proliferative and secretory phase of the menstrual cycle as well as in cultured HESCs. We also found that SRC-3 depletion in cultured HESCs results in a significant attenuation in the induction of a wide-range of established biomarkers of decidualization, despite exposure of these cells to a deciduogenic stimulus and normal progesterone receptor expression. These molecular findings are supported at the cellular level by the inability of HESCs to morphologically transform from a stromal fibroblastoid cell to an epithelioid decidual cell when endogenous SRC-3 levels are markedly reduced. To identify genes, signaling pathways and networks that are controlled by SRC-3 and potentially important for hormone-dependent decidualization, we performed RNA-sequencing on HESCs in which SRC-3 levels were significantly reduced at the time of administering the deciduogenic stimulus. Comparing HESC controls with HESCs deficient in SRC-3, gene enrichment analysis of the differentially expressed gene set revealed an overrepresentation of genes involved in chromatin remodeling, cell proliferation/motility, and programmed cell death. These predictive bioanalytic results were confirmed by the demonstration that SRC-3 is required for the expansion, migratory and invasive activities of the HESC population, cellular properties that are required in vivo in the formation or functioning of the decidua. Collectively, our results support SRC-3 as an important coregulator in HESC decidualization. Since perturbation of normal homeostatic levels of SRC-3 is linked with common gynecological disorders diagnosed in reproductive age women, this endometrial coregulator-along with its new molecular targets described here-may open novel clinical avenues in the diagnosis and/or treatment of a non-receptive endometrium, particularly in patients presenting non-aneuploid early pregnancy loss., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (© 2022 Maurya, Szwarc, Lonard, Gibbons, Wu, O'Malley, DeMayo and Lydon.)
- Published
- 2022
- Full Text
- View/download PDF
28. Spatial definition of the human progesterone receptor-B transcriptional complex.
- Author
-
Yu X, Yi P, Panigrahi AK, Lumahan LEV, Lydon JP, Lonard DM, Lutdke SJ, Wang Z, and O'Malley BW
- Abstract
We report the quaternary structure of core transcriptional complex for the full-length human progesterone receptor-B (PR-B) homodimer with primary coactivator steroid receptor coactivator-2 (SRC-2) and the secondary coactivator p300/CREB-binding protein (CBP). The PR-B homodimer engages one SRC-2 mainly through its activation function 1 (AF1) in N-terminus. SRC-2 is positioned between PR-B and p300 leaving space for direct interaction between PR-B and p300 through PR-B's C-terminal AF2 and its unique AF3. Direct AF3/p300 interaction provides long-desired structural insights into the known functional differences between PR-B and the PR-A isoform lacking AF3. We reveal the contributions of each AF and demonstrate their structural basis in forming the PR-B dimer interface and PR-B/coactivator complex. Comparison of the PR-B/coactivator complex with other steroid receptor (estrogen receptor and androgen receptor) complexes also shows that each receptor has its unique mechanism for recruiting coactivators due to the highly variable N-termini among receptors., Competing Interests: The authors declare no competing interests., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
29. Conditional knockout of leptin receptor in the female reproductive tract reduces fertility due to parturition defects in mice.
- Author
-
Pennington KA, Oestreich AK, Cataldo KH, Fogliatti CM, Lightner C, Lydon JP, and Schulz LC
- Subjects
- Animals, Embryo Implantation physiology, Female, Mice, Mice, Knockout, Parturition, Pregnancy, Uterus, Fertility genetics, Receptors, Leptin genetics
- Abstract
Leptin is required for fertility, including initiation of estrous cycles. It is therefore challenging to assess the role of leptin signaling during pregnancy. Although neuron-specific transgene approaches suggest that leptin signaling in the central nervous system is most important, experiments with pharmacologic inhibition of leptin in the uterus or global replacement of leptin during pregnancy suggest leptin signaling in the reproductive tract may be required. Here, conditional leptin receptor knockout (Lepr cKO) with a progesterone receptor-driven Cre recombinase was used to examine the importance of leptin signaling in pregnancy. Lepr cKO mice have almost no leptin receptor in uterus or cervix, and slightly reduced leptin receptor levels in corpus luteum. Estrous cycles and progesterone concentrations were not affected by Lepr cKO. Numbers of viable embryos did not differ between primiparous control and Lepr cKO dams on Days 6.5 and 17.5 of pregnancy, despite a slight reduction in the ratio of embryos to corpora lutea, showing that uterine leptin receptor signaling is not required for embryo implantation. Placentas of Lepr cKO dams had normal weight and structure. However, over four parities, Lepr cKO mice produced 22% fewer live pups than controls, and took more time from pairing to delivery by their fourth parity. Abnormal birth outcomes of either dystocia or dead pups occurred in 33% of Lepr cKO deliveries but zero control deliveries, and the average time to deliver each pup after crouching was significantly increased. Thus, leptin receptor signaling in the reproductive tract is required for normal labor and delivery., (Published by Oxford University Press on behalf of Society for the Study of Reproduction 2022.)
- Published
- 2022
- Full Text
- View/download PDF
30. Hippo-TAZ signaling is the master regulator of the onset of triple-negative basal-like breast cancers.
- Author
-
Soyama H, Nishio M, Otani J, Sakuma T, Takao S, Hara S, Masuda T, Mimori K, Toyokuni S, Lydon JP, Nakao K, Nishina H, Fukumoto T, Maehama T, and Suzuki A
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Carcinogenesis genetics, Cell Line, Tumor, Female, Gene Deletion, Humans, Intracellular Signaling Peptides and Proteins genetics, Mice, Receptors, Estrogen genetics, Transcriptional Coactivator with PDZ-Binding Motif Proteins, Tumor Suppressor Protein p53 genetics, YAP-Signaling Proteins genetics, Hippo Signaling Pathway genetics, Mammary Neoplasms, Experimental genetics, Precancerous Conditions genetics, Triple Negative Breast Neoplasms genetics
- Abstract
Breast cancer is the most frequent malignancy in women worldwide. Basal-like breast cancer (BLBC) is the most aggressive form of this disease, and patients have a poor prognosis. Here, we present data suggesting that the Hippo-transcriptional coactivator with PDZ-binding motif (TAZ) pathway is a key driver of BLBC onset and progression. Deletion of Mob1a/b in mouse mammary luminal epithelium induced rapid and highly reproducible mammary tumorigenesis that was dependent on TAZ but not yes-associated protein 1 (YAP1). In situ early-stage BLBC-like malignancies developed in mutant animals by 2 wk of age, and invasive BLBC appeared by 4 wk. In a human estrogen receptor
+ luminal breast cancer cell line, TAZ hyperactivation skewed the features of these luminal cells to the basal phenotype, consistent with the aberrant TAZ activation frequently observed in human precancerous BLBC lesions. TP53 mutation is rare in human precancerous BLBC but frequent in invasive BLBC. Addition of Trp53 deficiency to our Mob1a/b -deficient mouse model enhanced tumor grade and accelerated cancer progression. Our work justifies targeting the Hippo-TAZ pathway as a therapy for human BLBC, and our mouse model represents a powerful tool for evaluating candidate agents.- Published
- 2022
- Full Text
- View/download PDF
31. Early growth response 1 transcription factor is essential for the pathogenic properties of human endometriotic epithelial cells.
- Author
-
Maurya VK, Szwarc MM, Fernandez-Valdivia R, Lonard DM, Yong S, Joshi N, Fazleabas AT, and Lydon JP
- Subjects
- Cell Movement, Endometrium metabolism, Epithelial Cells metabolism, Female, Humans, Stromal Cells metabolism, Transcription Factors metabolism, Early Growth Response Protein 1 genetics, Endometriosis metabolism
- Abstract
Although a non-malignant gynecological disorder, endometriosis displays some pathogenic features of malignancy, such as cell proliferation, migration, invasion and adaptation to hypoxia. Current treatments of endometriosis include pharmacotherapy and/or surgery, which are of limited efficacy and often associated with adverse side effects. Therefore, to develop more effective therapies to treat this disease, a broader understanding of the underlying molecular mechanisms that underpin endometriosis needs to be attained. Using immortalized human endometriotic epithelial and stromal cell lines, we demonstrate that the early growth response 1 (EGR1) transcription factor is essential for cell proliferation, migration and invasion, which represent some of the pathogenic properties of endometriotic cells. Genome-wide transcriptomics identified an EGR1-dependent transcriptome in human endometriotic epithelial cells that potentially encodes a diverse spectrum of proteins that are known to be involved in tissue pathologies. To underscore the utility of this transcriptomic data set, we demonstrate that carbonic anhydrase 9 (CA9), a homeostatic regulator of intracellular pH, is not only a molecular target of EGR1 but is also important for maintaining many of the cellular properties of human endometriotic epithelial cells that are also ascribed to EGR1. Considering therapeutic intervention strategies are actively being developed for EGR1 and CAIX in the treatment of other pathologies, we believe EGR1 and its transcriptome (which includes CA9) will offer not only a new conceptual framework to advance our understanding of endometriosis but will also furnish new molecular vulnerabilities to be leveraged as potential therapeutic options in the future treatment of endometriosis.
- Published
- 2022
- Full Text
- View/download PDF
32. Corrigendum: Nuclear Progesterone Receptor Expressed by the Cortical Thymic Epithelial Cells Dictates Thymus Involution in Murine Pregnancy.
- Author
-
Ahn SH, Nguyen SL, Kim TH, Jeong JW, Arora R, Lydon JP, and Petroff MG
- Abstract
[This corrects the article DOI: 10.3389/fendo.2022.846226.]., (Copyright © 2022 Ahn, Nguyen, Kim, Jeong, Arora, Lydon and Petroff.)
- Published
- 2022
- Full Text
- View/download PDF
33. Aberrant uterine folding in mice disrupts implantation chamber formation and alignment of embryo-uterine axes.
- Author
-
Madhavan MK, DeMayo FJ, Lydon JP, Joshi NR, Fazleabas AT, and Arora R
- Subjects
- Animals, Embryo, Mammalian, Epithelium, Female, Horses, Humans, Mammals, Mice, Pregnancy, Embryo Implantation, Uterus
- Abstract
The uterine luminal epithelium folds characteristically in mammals, including humans, horses and rodents. Improper uterine folding in horses results in pregnancy failure, but the precise function of folds remains unknown. Here, we uncover dynamic changes in the 3D uterine folding pattern during early pregnancy with the entire lumen forming pre-implantation transverse folds along the mesometrial-antimesometrial axis. Using a time course, we show that transverse folds are formed before embryo spacing, whereas implantation chambers form as the embryo begins attachment. Thus, folds and chambers are two distinct structures. Transverse folds resolve to form a flat implantation region, after which an embryo arrives at its center to attach and form the post-implantation chamber. Our data also suggest that the implantation chamber facilitates embryo rotation and its alignment along the uterine mesometrial-antimesometrial axis. Using WNT5A- and RBPJ-deficient mice that display aberrant folds, we show that embryos trapped in longitudinal folds display misalignment of the embryo-uterine axes, abnormal chamber formation and defective post-implantation morphogenesis. These mouse models with disrupted uterine folding provide an opportunity to understand uterine structure-based mechanisms that are crucial for implantation and pregnancy success. This article has an associated 'The people behind the papers' interview., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
34. Intraovarian, Isoform-Specific Transcriptional Roles of Progesterone Receptor in Ovulation.
- Author
-
Smith KM, Dinh DT, Akison LK, Nicholls M, Dunning KR, Morimoto A, Lydon JP, Russell DL, and Robker RL
- Subjects
- Animals, Female, Granulosa Cells metabolism, Mammals metabolism, Mice, Mice, Knockout, Progesterone pharmacology, Protein Isoforms metabolism, Ovulation, Receptors, Progesterone genetics, Receptors, Progesterone metabolism
- Abstract
Progesterone receptor (PGR) activity is obligatory for mammalian ovulation; however, there is no established direct functional pathway explaining how progesterone receptor completely and specifically regulates oocyte release. This study examined the overarching cell- and isoform-specific effects of the PGR within each cellular compartment of the ovary, using mice null for the PGR (PRKO), as well as isoform-specific null mice. The PGR was expressed in ovarian granulosa and stromal cells and although PRKO ovaries showed no visible histological changes in preovulatory ovarian morphology, follicle rupture did not occur. Reciprocal ovarian transplant experiments established the necessity of ovarian PGR expression for ovulation. Cumulus-oocyte complexes of PRKO mice exhibited normal morphology but showed some altered gene expression. The examination of mitochondrial activity showed subtle differences in PRKO oocytes but no differences in granulosa cell respiration, glycolysis or β-oxidation. Concurrently, RNA-seq identified novel functional pathways through which the PGR may regulate ovulation. PGR-A was the predominant transcriptionally active isoform in granulosa cells and 154 key PGR-dependent genes were identified, including a secondary network of transcription factors. In addition, the PGR regulated unique gene networks in the ovarian stroma. Collectively, we establish the effector pathways activated by the PGR across the ovarian cell types and conclude that PGR coordinates gene expression in the cumulus, granulosa and stromal cells at ovulation. Identifying these networks linking the PGR to ovulation provides novel targets for fertility therapeutics and nonhormonal contraceptive development.
- Published
- 2022
- Full Text
- View/download PDF
35. Nuclear Progesterone Receptor Expressed by the Cortical Thymic Epithelial Cells Dictates Thymus Involution in Murine Pregnancy.
- Author
-
Ahn SH, Nguyen SL, Kim TH, Jeong JW, Arora R, Lydon JP, and Petroff MG
- Subjects
- Animals, Epithelial Cells metabolism, Female, Mice, Mice, Transgenic, Pregnancy, Uterus metabolism, Progesterone metabolism, Receptors, Progesterone genetics, Receptors, Progesterone metabolism
- Abstract
Progesterone is a gonadal pro-gestational hormone that is absolutely necessary for the success of pregnancy. Most notable actions of progesterone are observed in the female reproductive organs, the uterus and the ovary. Acting through the nuclear progesterone receptor (PGR), progesterone prepares the endometrium for implantation of the embryo. Interestingly, the maternal thymus also is a known expressor of Pgr ; its absence is associated with murine pregnancy complications. However, the localization of its expression and its functional importance were not known. Here, we used a transgenic dual fluorescent reporter mouse model and genetic deletion of Pgr in Foxn1+ thymic epithelial cells (TEC) to demonstrate TEC-specific Pgr expression in pregnancy, especially in the cortex where thymocyte maturation occurs. Using our TEC-specific Pgr deletion mouse model, we demonstrate that TEC-specific Pgr is necessary for pregnancy-induced thymic involution in pregnancy. Our investigation reveals that PGR expression is upregulated in the cortical thymic epithelial cells during pregnancy, and that PGR expression is important for thymic involution during murine pregnancy., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The reviewer S-PW declared a past collaboration with two of the authors JL and J-WJ to the handling editor., (Copyright © 2022 Ahn, Nguyen, Kim, Jeong, Arora, Lydon and Petroff.)
- Published
- 2022
- Full Text
- View/download PDF
36. EZH2 and Endometrial Cancer Development: Insights from a Mouse Model.
- Author
-
Fang X, Ni N, Wang X, Tian Y, Ivanov I, Rijnkels M, Bayless KJ, Lydon JP, and Li Q
- Subjects
- Animals, Carcinogenesis pathology, Disease Models, Animal, Female, Humans, Mice, Polycomb Repressive Complex 2 genetics, Uterus pathology, Endometrial Neoplasms genetics, Endometrial Neoplasms pathology, Enhancer of Zeste Homolog 2 Protein genetics
- Abstract
Enhancer of zeste homolog 2 (EZH2), a core component of polycomb repressive complex 2, plays an important role in cancer development. As both oncogenic and tumor suppressive functions of EZH2 have been documented in the literature, the objective of this study is to determine the impact of Ezh2 deletion on the development and progression of endometrial cancer induced by inactivation of phosphatase and tensin homolog ( PTEN ), a tumor suppressor gene frequently dysregulated in endometrial cancer patients. To this end, we created mice harboring uterine deletion of both Ezh2 and Pten using Cre recombinase driven by the progesterone receptor ( Pgr ) promoter. Our results showed reduced tumor burden in Pten
d/d ; Ezh2d/d mice compared with that of Ptend/d mice during early carcinogenesis. The decreased Ki67 index in EZH2 and PTEN-depleted uteri versus that in PTEN-depleted uteri indicated an oncogenic role of EZH2 during early tumor development. However, mice harboring uterine deletion of both Ezh2 and Pten developed unfavorable disease outcome, accompanied by exacerbated epithelial stratification and heightened inflammatory response. The observed effect was non-cell autonomous and mediated by altered immune response evidenced by massive accumulation of intraluminal neutrophils, a hallmark of endometrial carcinoma in Ptend/d ; Ezh2d/d mice during disease progression. Hence, these results reveal dual roles of EZH2 in endometrial cancer development.- Published
- 2022
- Full Text
- View/download PDF
37. Deficiency of PARP-1 and PARP-2 in the mouse uterus results in decidualization failure and pregnancy loss.
- Author
-
Kelleher AM, Setlem R, Dantzer F, DeMayo FJ, Lydon JP, and Kraus WL
- Subjects
- Animals, Embryo Implantation physiology, Embryo, Mammalian metabolism, Female, Mice, Mice, Inbred C57BL, Mice, Knockout, Pregnancy, Pregnancy Outcome, Signal Transduction physiology, Stromal Cells metabolism, Abortion, Spontaneous metabolism, Decidua metabolism, Poly (ADP-Ribose) Polymerase-1 metabolism, Poly(ADP-ribose) Polymerases metabolism, Uterus metabolism
- Abstract
Miscarriage is a common complication of pregnancy for which there are few clinical interventions. Deficiency in endometrial stromal cell decidualization is considered a major contributing factor to pregnancy loss; however, our understanding of the underlying mechanisms of decidual deficiency are incomplete. ADP ribosylation by PARP-1 and PARP-2 has been linked to physiological processes essential to successful pregnancy outcomes. Here, we report that the catalytic inhibition or genetic ablation of PARP-1 and PARP-2 in the uterus lead to pregnancy loss in mice. Notably, the absence of PARP-1 and PARP-2 resulted in increased p53 signaling and an increased population of senescent decidual cells. Molecular and histological analysis revealed that embryo attachment and the removal of the luminal epithelium are not altered in uterine Parp1 , Parp2 knockout mice, but subsequent decidualization failure results in pregnancy loss. These findings provide evidence for a previously unknown function of PARP-1 and PARP-2 in mediating decidualization for successful pregnancy establishment., Competing Interests: Competing interest statement: W.L.K. is a founder, consultant, and Scientific Advisory Board member for Ribon Therapeutics, Inc. and ARase Therapeutics, Inc. He is also coholder of US Patent 9,599,606 covering the ADP-ribose detection reagent used herein, which has been licensed to and is sold by EMD Millipore.
- Published
- 2021
- Full Text
- View/download PDF
38. Vaginal Squamous Cell Carcinoma Develops in Mice with Conditional Arid1a Loss and Gain of Oncogenic Kras Driven by Progesterone Receptor Cre.
- Author
-
Wang X, Praça MSL, Wendel JRH, Emerson RE, DeMayo FJ, Lydon JP, and Hawkins SM
- Subjects
- Animals, Carcinoma, Squamous Cell pathology, Disease Progression, Female, Integrases, Mice, Squamous Intraepithelial Lesions genetics, Squamous Intraepithelial Lesions pathology, Vaginal Neoplasms pathology, Carcinoma, Squamous Cell genetics, DNA-Binding Proteins genetics, Disease Models, Animal, Proto-Oncogene Proteins p21(ras) genetics, Receptors, Progesterone genetics, Transcription Factors genetics, Vaginal Neoplasms genetics
- Abstract
Oncogenic KRAS mutations are a common finding in endometrial cancers. Recent sequencing studies indicate that loss-of-function mutations in the ARID1A gene are enriched in gynecologic malignant tumors. However, neither of these genetic insults alone are sufficient to develop gynecologic cancer. To determine the role of the combined effects of deletion of Arid1a and oncogenic Kras, Arid1a
flox/flox mice were crossed with KrasLox-Stop-Lox-G12D/+ mice using progesterone receptor Cre (PgrCre/+ ). Histologic analysis and immunohistochemistry of survival studies were used to characterize the mutant mouse phenotype. Hormone dependence was evaluated by ovarian hormone depletion and estradiol replacement. Arid1aflox/flox ; KrasLox-Stop-Lox-G12D/+ ; PgrCre/+ mice were euthanized early because of invasive vaginal squamous cell carcinoma. Younger mice had precancerous intraepithelial lesions. Immunohistochemistry supported the pathological diagnosis with abnormal expression and localization of cytokeratin 5, tumor protein P63, cyclin-dependent kinase inhibitor 2A, and Ki-67, the marker of proliferation. Ovarian hormone deletion in Arid1aflox/flox ; KrasLox-Stop-Lox-G12D/+ ; PgrCre/+ mice resulted in atrophic vaginal epithelium without evidence of vaginal tumors. Estradiol replacement in ovarian hormone-depleted Arid1aflox/flox ; KrasLox-Stop-Lox-G12D/+ ; PgrCre/+ mice resulted in lesions that resembled the squamous cell carcinoma in intact mice. Therefore, this mouse can be used to study the transition from benign precursor lesions into invasive vaginal human papillomavirus-independent squamous cell carcinoma, offering insights into progression and pathogenesis of this rare disease., (Copyright © 2021 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
39. Cell-type specific analysis of physiological action of estrogen in mouse oviducts.
- Author
-
McGlade EA, Herrera GG, Stephens KK, Olsen SLW, Winuthayanon S, Guner J, Hewitt SC, Korach KS, DeMayo FJ, Lydon JP, Monsivais D, and Winuthayanon W
- Subjects
- Animals, Estrogens pharmacology, Fallopian Tubes cytology, Fallopian Tubes drug effects, Female, Gene Expression Profiling, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Oviducts cytology, Oviducts drug effects, Receptors, Progesterone physiology, Biomarkers metabolism, Estradiol pharmacology, Fallopian Tubes physiology, Gene Expression Regulation, Developmental drug effects, Insulin-Like Growth Factor I physiology, Oviducts physiology, Single-Cell Analysis methods
- Abstract
One of the endogenous estrogens, 17β-estradiol (E
2 ) is a female steroid hormone secreted from the ovary. It is well established that E2 causes biochemical and histological changes in the uterus. However, it is not completely understood how E2 regulates the oviductal environment in vivo. In this study, we assessed the effect of E2 on each oviductal cell type, using an ovariectomized-hormone-replacement mouse model, single-cell RNA-sequencing (scRNA-seq), in situ hybridization, and cell-type-specific deletion in mice. We found that each cell type in the oviduct responded to E2 distinctively, especially ciliated and secretory epithelial cells. The treatment of exogenous E2 did not drastically alter the transcriptomic profile from that of endogenous E2 produced during estrus. Moreover, we have identified and validated genes of interest in our datasets that may be used as cell- and region-specific markers in the oviduct. Insulin-like growth factor 1 (Igf1) was characterized as an E2 -target gene in the mouse oviduct and was also expressed in human fallopian tubes. Deletion of Igf1 in progesterone receptor (Pgr)-expressing cells resulted in female subfertility, partially due to an embryo developmental defect and embryo retention within the oviduct. In summary, we have shown that oviductal cell types, including epithelial, stromal, and muscle cells, are differentially regulated by E2 and support gene expression changes, such as growth factors that are required for normal embryo development and transport in mouse models. Furthermore, we have identified cell-specific and region-specific gene markers for targeted studies and functional analysis in vivo., (© 2021 Federation of American Societies for Experimental Biology.)- Published
- 2021
- Full Text
- View/download PDF
40. Illuminating the "Black Box" of Progesterone-Dependent Embryo Implantation Using Engineered Mice.
- Author
-
Maurya VK, DeMayo FJ, and Lydon JP
- Abstract
Synchrony between progesterone-driven endometrial receptivity and the arrival of a euploid blastocyst is essential for embryo implantation, a prerequisite event in the establishment of a successful pregnancy. Advancement of embryo implantation within the uterus also requires stromal fibroblasts of the endometrium to transform into epithelioid decidual cells, a progesterone-dependent cellular transformation process termed decidualization. Although progesterone is indispensable for these cellular processes, the molecular underpinnings are not fully understood. Because human studies are restricted, much of our fundamental understanding of progesterone signaling in endometrial periimplantation biology comes from in vitro and in vivo experimental systems. In this review, we focus on the tremendous progress attained with the use of engineered mouse models together with high throughput genome-scale analysis in disclosing key signals, pathways and networks that are required for normal endometrial responses to progesterone during the periimplantation period. Many molecular mediators and modifiers of the progesterone response are implicated in cross talk signaling between epithelial and stromal cells of the endometrium, an intercellular communication system that is critical for the ordered spatiotemporal control of embryo invasion within the maternal compartment. Accordingly, derailment of these signaling systems is causally linked with infertility, early embryo miscarriage and gestational complications that symptomatically manifest later in pregnancy. Such aberrant progesterone molecular responses also contribute to endometrial pathologies such as endometriosis, endometrial hyperplasia and cancer. Therefore, our review makes the case that further identification and functional analysis of key molecular mediators and modifiers of the endometrial response to progesterone will not only provide much-needed molecular insight into the early endometrial cellular changes that promote pregnancy establishment but lend credible hope for the development of more effective mechanism-based molecular diagnostics and precision therapies in the clinical management of female infertility, subfertility and a subset of gynecological morbidities., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Maurya, DeMayo and Lydon.)
- Published
- 2021
- Full Text
- View/download PDF
41. Progesterone receptor isoform B regulates the Oxtr - Plcl2 - Trpc3 pathway to suppress uterine contractility.
- Author
-
Peavey MC, Wu SP, Li R, Liu J, Emery OM, Wang T, Zhou L, Wetendorf M, Yallampalli C, Gibbons WE, Lydon JP, and DeMayo FJ
- Subjects
- Animals, Female, Mice, Mice, Mutant Strains, Parturition physiology, Pregnancy, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, Transcriptome, Gene Expression Regulation physiology, Intracellular Signaling Peptides and Proteins genetics, Receptors, Oxytocin genetics, Receptors, Progesterone physiology, TRPC Cation Channels genetics, Uterine Contraction genetics
- Abstract
Uterine contractile dysfunction leads to pregnancy complications such as preterm birth and labor dystocia. In humans, it is hypothesized that progesterone receptor isoform PGR-B promotes a relaxed state of the myometrium, and PGR-A facilitates uterine contraction. This hypothesis was tested in vivo using transgenic mouse models that overexpress PGR-A or PGR-B in smooth muscle cells. Elevated PGR-B abundance results in a marked increase in gestational length compared to control mice (21.1 versus 19.1 d respectively, P < 0.05). In both ex vivo and in vivo experiments, PGR-B overexpression leads to prolonged labor, a significant decrease in uterine contractility, and a high incidence of labor dystocia. Conversely, PGR-A overexpression leads to an increase in uterine contractility without a change in gestational length. Uterine RNA sequencing at midpregnancy identified 1,174 isoform-specific downstream targets and 424 genes that are commonly regulated by both PGR isoforms. Gene signature analyses further reveal PGR-B for muscle relaxation and PGR-A being proinflammatory. Elevated PGR-B abundance reduces Oxtr and Trpc3 and increases Plcl2 expression, which manifests a genetic profile of compromised oxytocin signaling. Functionally, both endogenous PLCL2 and its paralog PLCL1 can attenuate uterine muscle cell contraction in a CRISPRa-based assay system. These findings provide in vivo support that PGR isoform levels determine distinct transcriptomic landscapes and pathways in myometrial function and labor, which may help further the understanding of abnormal uterine function in the clinical setting., Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
42. Different Cre systems induce differential microRNA landscapes and abnormalities in the female reproductive tracts of Dgcr8 conditional knockout mice.
- Author
-
Kim YS, Yang SC, Park M, Choi Y, DeMayo FJ, Lydon JP, Kim HR, Lim HJ, and Song H
- Subjects
- Animals, Female, Integrases metabolism, Integrases pharmacology, Mice, Knockout, MicroRNAs genetics, Oviducts growth & development, Oviducts metabolism, RNA-Binding Proteins metabolism, Reproduction physiology, Uterus metabolism, Mice, RNA-Binding Proteins genetics, Reproduction genetics, Uterus pathology
- Abstract
Objectives: The female reproductive tract comprises several different cell types. Using three representative Cre systems, we comparatively analysed the phenotypes of Dgcr8 conditional knockout (cKO) mice to understand the function of Dgcr8, involved in canonical microRNA biogenesis, in the female reproductive tract., Materials and Methods: Dgcr8
f/f mice were crossed with Ltficre/+ , Amhr2cre/+ or PRcre/+ mice to produce mice deficient in Dgcr8 in epithelial (Dgcr8ed/ed ), mesenchymal (Dgcr8md/md ) and all the compartments (Dgcr8td/td ) in the female reproductive tract. Reproductive phenotypes were evaluated in Dgcr8 cKO mice. Uteri and/or oviducts were used for small RNA-seq, mRNA-seq, real-time RT-PCR, and/or morphologic and histological analyses., Result: Dgcr8ed/ed mice did not exhibit any distinct defects, whereas Dgcr8md/md mice showed sub-fertility and oviductal smooth muscle deformities. Dgcr8td/td mice were infertile due to anovulation and acute inflammation in the female reproductive tract and suffered from an atrophic uterus with myometrial defects. The microRNAs and mRNAs related to immune modulation and/or smooth muscle growth were systemically altered in the Dgcr8td/td uterus. Expression profiles of dysregulated microRNAs and mRNAs in the Dgcr8td/td uterus were different from those in other genotypes in a Cre-dependent manner., Conclusions: Dgcr8 deficiency with different Cre systems induces overlapping but distinct phenotypes as well as the profiles of microRNAs and their target mRNAs in the female reproductive tract, suggesting the importance of selecting the appropriate Cre driver to investigate the genes of interest., (© 2021 The Authors. Cell Proliferation Published by John Wiley & Sons Ltd.)- Published
- 2021
- Full Text
- View/download PDF
43. Targeting progesterone signaling prevents metastatic ovarian cancer.
- Author
-
Kim O, Park EY, Kwon SY, Shin S, Emerson RE, Shin YH, DeMayo FJ, Lydon JP, Coffey DM, Hawkins SM, Quilliam LA, Cheon DJ, Fernández FM, Nephew KP, Karpf AR, Widschwendter M, Sood AK, Bast RC Jr, Godwin AK, Miller KD, Cho CH, and Kim J
- Subjects
- Adult, Animals, Breast pathology, Breast Neoplasms genetics, Breast Neoplasms pathology, Breast Neoplasms prevention & control, Cystadenocarcinoma, Serous chemistry, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous pathology, Disease Models, Animal, Estradiol administration & dosage, Female, Humans, Mice, Middle Aged, Mifepristone therapeutic use, Mutation, Neoplasms, Experimental chemically induced, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Neoplasms, Experimental prevention & control, Ovarian Neoplasms chemically induced, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Ovary pathology, Ovary surgery, Progesterone administration & dosage, Progesterone metabolism, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, Salpingo-oophorectomy, Signal Transduction drug effects, Signal Transduction genetics, BRCA1 Protein genetics, Cystadenocarcinoma, Serous prevention & control, Mifepristone pharmacology, Ovarian Neoplasms prevention & control, Progesterone antagonists & inhibitors
- Abstract
Effective cancer prevention requires the discovery and intervention of a factor critical to cancer development. Here we show that ovarian progesterone is a crucial endogenous factor inducing the development of primary tumors progressing to metastatic ovarian cancer in a mouse model of high-grade serous carcinoma (HGSC), the most common and deadliest ovarian cancer type. Blocking progesterone signaling by the pharmacologic inhibitor mifepristone or by genetic deletion of the progesterone receptor (PR) effectively suppressed HGSC development and its peritoneal metastases. Strikingly, mifepristone treatment profoundly improved mouse survival (∼18 human years). Hence, targeting progesterone/PR signaling could offer an effective chemopreventive strategy, particularly in high-risk populations of women carrying a deleterious mutation in the BRCA gene., Competing Interests: Competing interest statement: J.K. and K.D.M. are listed as inventors on patent applications filed by Indiana University related to targeting progesterone signaling.
- Published
- 2020
- Full Text
- View/download PDF
44. Transforming growth factor beta signaling and decidual integrity in mice†.
- Author
-
Fang X, Ni N, Gao Y, Lydon JP, Ivanov I, Rijnkels M, Bayless KJ, and Li Q
- Subjects
- Animals, Bone Morphogenetic Proteins genetics, Bone Morphogenetic Proteins metabolism, Cell Proliferation, Endometrium physiology, Extracellular Signal-Regulated MAP Kinases genetics, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Gene Expression Regulation, Developmental, MAP Kinase Signaling System, Mice, Mice, Knockout, Receptor, Transforming Growth Factor-beta Type I genetics, Signal Transduction, Transforming Growth Factor beta genetics, Up-Regulation, Uterus, Gene Expression Regulation physiology, Receptor, Transforming Growth Factor-beta Type I metabolism, Transforming Growth Factor beta metabolism
- Abstract
Transforming growth factor beta (TGFβ) signaling regulates multifaceted reproductive processes. It has been shown that the type 1 receptor of TGFβ (TGFBR1) is indispensable for female reproductive tract development, implantation, placental development, and fertility. However, the role of TGFβ signaling in decidual development and function remains poorly defined. Our objective is to determine the impact of uterine-specific deletion of Tgfbr1 on decidual integrity, with a focus on the cellular and molecular properties of the decidua during development. Our results show that the developmental dynamics of the decidua is altered in TGFBR1 conditionally depleted uteri from embryonic day (E) 5.5 to E8.5, substantiated by downregulation of genes associated with inflammatory responses and uterine natural killer cell abundance, reduced presence of nondecidualized fibroblasts in the antimesometrial region, and altered decidual cell development. Notably, conditional ablation of TGFBR1 results in the formation of decidua containing more abundant alpha smooth muscle actin (ACTA2)-positive cells at the peripheral region of the antimesometrial side versus controls at E6.5-E8.5. This finding is corroborated by upregulation of a subset of smooth muscle marker genes in Tgfbr1 conditionally deleted decidua at E6.5 and E8.5. Moreover, increased cell proliferation and enhanced decidual ERK1/2 signaling were found in Tgfbr1 conditional knockout mice upon decidual regression. In summary, conditional ablation of TGFBR1 in the uterus profoundly impacts the cellular and molecular properties of the decidua. Our results suggest that TGFBR1 in uterine epithelial and stromal compartments is important for the integrity of the decidua, a transient but crucial structure that supports embryo development., (© The Author(s) 2020. Published by Oxford University Press on behalf of Society for the Study of Reproduction. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
45. WNK1 regulates uterine homeostasis and its ability to support pregnancy.
- Author
-
Chi RA, Wang T, Huang CL, Wu SP, Young SL, Lydon JP, and DeMayo FJ
- Subjects
- Animals, Female, Forkhead Box Protein O1 genetics, Gene Regulatory Networks, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Phosphorylation, Pregnancy, Protein Phosphatase 2 genetics, Proteome, Proto-Oncogene Proteins c-akt genetics, Transcriptome, Forkhead Box Protein O1 metabolism, Homeostasis, Protein Phosphatase 2 metabolism, Proto-Oncogene Proteins c-akt metabolism, Reproduction, Uterus physiology, WNK Lysine-Deficient Protein Kinase 1 physiology
- Abstract
WNK1 (with no lysine [K] kinase 1) is an atypical kinase protein ubiquitously expressed in humans and mice. A mutation in its encoding gene causes hypertension in humans, which is associated with abnormal ion homeostasis. WNK1 is critical for in vitro decidualization in human endometrial stromal cells, thereby demonstrating its importance in female reproduction. Using a mouse model, WNK1 was ablated in the female reproductive tract to define its in vivo role in uterine biology. Loss of WNK1 altered uterine morphology, causing endometrial epithelial hyperplasia, adenomyotic features, and a delay in embryo implantation, ultimately resulting in compromised fertility. Combining transcriptomic, proteomic, and interactomic analyses revealed a potentially novel regulatory pathway whereby WNK1 represses AKT phosphorylation through protein phosphatase 2A (PP2A) in endometrial cells from both humans and mice. We show that WNK1 interacted with PPP2R1A, the alpha isoform of the PP2A scaffold subunit. This maintained the levels of PP2A subunits and stabilized its activity, which then dephosphorylated AKT. Therefore, loss of WNK1 reduced PP2A activity, causing AKT hypersignaling. Using FOXO1 as a readout of AKT activity, we demonstrate that there was escalated FOXO1 phosphorylation and nuclear exclusion, leading to a disruption in the expression of genes that are crucial for embryo implantation.
- Published
- 2020
- Full Text
- View/download PDF
46. Increased FOXL2 expression alters uterine structures and functions†.
- Author
-
Li R, Wu SP, Zhou L, Nicol B, Lydon JP, Yao HH, and DeMayo FJ
- Subjects
- Animals, Endometrium metabolism, Female, Forkhead Box Protein L2 genetics, Gene Expression Regulation, Mice, Mice, Transgenic, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, Signal Transduction physiology, Transcriptome, Urogenital Abnormalities genetics, Forkhead Box Protein L2 metabolism, Urogenital Abnormalities metabolism, Uterus abnormalities, Uterus metabolism
- Abstract
The transcription factor forkhead box L2 (FOXL2) regulates sex differentiation and reproductive function. Elevated levels of this transcription factor have been observed in the diseases of the uterus, such as endometriosis. However, the impact of elevated FOXL2 expression on uterine physiology remains unknown. In order to determine the consequences of altered FOXL2 in the female reproductive axis, we generated mice with over-expression of FOXL2 (FOXL2OE) by crossing Foxl2LsL/+ with the Progesterone receptor Pgrcre model. FOXL2OE uterus showed severe morphological abnormality including abnormal epithelial stratification, blunted adenogenesis, increased endometrial fibrosis, and disrupted myometrial morphology. In contrast, increasing FOXL2 levels specifically in uterine epithelium by crossing the Foxl2LsL/+ with the lactoferrin Ltficre mice resulted in the eFOXL2OE mice with uterine epithelial stratification but without defects in endometrial fibrosis and adenogenesis, demonstrating a role of the endometrial stroma in the uterine abnormalities of the FOXL2OE mice. Transcriptomic analysis of 12 weeks old Pgrcre and FOXL2OE uterus at diestrus stage showed multiple signaling pathways related with cellular matrix, wnt/β-catenin, and altered cell cycle. Furthermore, we found FOXL2OE mice were sterile. The infertility was caused in part by a disruption of the hypophyseal ovarian axis resulting in an anovulatory phenotype. The FOXL2OE mice failed to show decidual responses during artificial decidualization in ovariectomized mice demonstrating the uterine contribution to the infertility phenotype. These data support that aberrantly increased FOXL2 expressions in the female reproductive tract can disrupt ovarian and uterine functions., (Published by Oxford University Press on behalf of Society for the Study of Reproduction 2020.)
- Published
- 2020
- Full Text
- View/download PDF
47. Constitutive expression of progesterone receptor isoforms promotes the development of hormone-dependent ovarian neoplasms.
- Author
-
Wetendorf M, Li R, Wu SP, Liu J, Creighton CJ, Wang T, Janardhan KS, Willson CJ, Lanz RB, Murphy BD, Lydon JP, and DeMayo FJ
- Subjects
- Animals, Cell Proliferation genetics, Disease Models, Animal, Estradiol blood, Estradiol metabolism, Female, Hormones blood, Humans, Mice, Knockout, Mice, Transgenic, Microscopy, Fluorescence, Ovarian Neoplasms metabolism, Progesterone blood, Progesterone metabolism, Receptors, Progesterone metabolism, Gene Expression Profiling methods, Hormones metabolism, Ovarian Neoplasms genetics, Receptors, Progesterone genetics, Transcriptome genetics
- Abstract
Differences in the relative abundances of the progesterone receptor (PGR) isoforms PGRA and PGRB are often observed in women with reproductive tract cancers. To assess the importance of the PGR isoform ratio in the maintenance of the reproductive tract, we generated mice that overexpress PGRA or PGRB in all PGR-positive tissues. Whereas few PGRA-overexpressing mice developed reproductive tract tumors, all PGRB-overexpressing mice developed ovarian neoplasms that were derived from ovarian luteal cells. Transcriptomic analyses of the ovarian tumors from PGRB-overexpressing mice revealed enhanced AKT signaling and a gene expression signature similar to those of human ovarian and endometrial cancers. Treating PGRB-overexpressing mice with the PGR antagonist RU486 stalled tumor growth and decreased the expression of cell cycle-associated genes, indicating that tumor growth and cell proliferation were hormone dependent in addition to being isoform dependent. Analysis of the PGRB cistrome identified binding events at genes encoding proteins that are critical regulators of mitotic phase entry. This work suggests a mechanism whereby an increase in the abundance of PGRB relative to that of PGRA drives neoplasia in vivo by stimulating cell cycling., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
48. Pten and Dicer1 loss in the mouse uterus causes poorly differentiated endometrial adenocarcinoma.
- Author
-
Wang X, Wendel JRH, Emerson RE, Broaddus RR, Creighton CJ, Rusch DB, Buechlein A, DeMayo FJ, Lydon JP, and Hawkins SM
- Subjects
- Adenocarcinoma, Clear Cell genetics, Animals, Cell Line, Tumor, DEAD-box RNA Helicases metabolism, Disease Models, Animal, Endometrial Neoplasms genetics, Endometrium pathology, Female, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Gene Knockout Techniques, Humans, Lim Kinases genetics, Mice, Mice, Transgenic, MicroRNAs metabolism, PTEN Phosphohydrolase metabolism, RNA-Seq, Ribonuclease III metabolism, Adenocarcinoma, Clear Cell pathology, Cell Differentiation genetics, DEAD-box RNA Helicases genetics, Endometrial Neoplasms pathology, PTEN Phosphohydrolase genetics, Ribonuclease III genetics
- Abstract
Endometrial cancer remains the most common gynecological malignancy in the United States. While the loss of the tumor suppressor, PTEN (phosphatase and tensin homolog), is well studied in endometrial cancer, recent studies suggest that DICER1, the endoribonuclease responsible for miRNA genesis, also plays a significant role in endometrial adenocarcinoma. Conditional uterine deletion of Dicer1 and Pten in mice resulted in poorly differentiated endometrial adenocarcinomas, which expressed Napsin A and HNF1B (hepatocyte nuclear factor 1 homeobox B), markers of clear-cell adenocarcinoma. Adenocarcinomas were hormone-independent. Treatment with progesterone did not mitigate poorly differentiated adenocarcinoma, nor did it affect adnexal metastasis. Transcriptomic analyses of DICER1 deleted uteri or Ishikawa cells revealed unique transcriptomic profiles and global miRNA downregulation. Computational integration of miRNA with mRNA targets revealed deregulated let-7 and miR-16 target genes, similar to published human DICER1-mutant endometrial cancers from TCGA (The Cancer Genome Atlas). Similar to human endometrial cancers, tumors exhibited dysregulation of ephrin-receptor signaling and transforming growth factor-beta signaling pathways. LIM kinase 2 (LIMK2), an essential molecule in p21 signal transduction, was significantly upregulated and represents a novel mechanism for hormone-independent pathogenesis of endometrial adenocarcinoma. This preclinical mouse model represents the first genetically engineered mouse model of poorly differentiated endometrial adenocarcinoma.
- Published
- 2020
- Full Text
- View/download PDF
49. Endogenous YAP1 activation drives immediate onset of cervical carcinoma in situ in mice.
- Author
-
Nishio M, To Y, Maehama T, Aono Y, Otani J, Hikasa H, Kitagawa A, Mimori K, Sasaki T, Nishina H, Toyokuni S, Lydon JP, Nakao K, Wah Mak T, Kiyono T, Katabuchi H, Tashiro H, and Suzuki A
- Subjects
- Animals, Carcinoma virology, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell virology, Cell Line, Cell Line, Tumor, Epithelial Cells metabolism, Epithelial Cells virology, Estrogens metabolism, Humans, Mice, Mice, Knockout, Oncogene Proteins, Viral metabolism, Papillomaviridae metabolism, Papillomaviridae pathogenicity, Papillomavirus E7 Proteins metabolism, Phosphatidylinositol 3-Kinase metabolism, Protein Tyrosine Phosphatases, Non-Receptor metabolism, Repressor Proteins metabolism, Root Caries virology, Signal Transduction physiology, Tumor Suppressor Protein p53 metabolism, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing metabolism, Carcinoma metabolism, Cell Cycle Proteins metabolism, Root Caries metabolism
- Abstract
Cervical cancer (CC) is usually initiated by infection with high-risk types of human papillomavirus (HPV). The HPV E6 and E7 proteins target p53 and RB, respectively, but other cellular targets likely exist. We generated uterus-specific MOB1A/B double KO (uMob1DKO) mice, which immediately developed cervical squamous cell carcinoma in situ. Mutant cervical epithelial cells showed YAP1-dependent hyperproliferation, altered self-renewal, impaired contact inhibition, and chromosomal instability. p53 activation was increased in uMob1DKO cells, and additional p53 loss in uMob1DKO mice accelerated tumor invasion. In human CC, strong YAP1 activation was observed from the precancerous stage. Human cells overexpressing HPV16 E6/E7 showed inactivation of not only p53 and RB but also PTPN14, boosting YAP1 activation. Estrogen, cigarette smoke condensate, and PI3K hyperactivation all increased YAP1 activity in human cervical epithelial cells, and PTPN14 depletion along with PI3K activation or estrogen treatment further enhanced YAP1. Thus, immediate CC onset may initiate when YAP1 activity exceeds an oncogenic threshold, making Hippo-YAP1 signaling a major CC driver., (© 2020 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.)
- Published
- 2020
- Full Text
- View/download PDF
50. 90 YEARS OF PROGESTERONE: New insights into progesterone receptor signaling in the endometrium required for embryo implantation.
- Author
-
DeMayo FJ and Lydon JP
- Subjects
- Animals, Female, Humans, Models, Biological, Receptors, Progesterone chemistry, Embryo Implantation, Endometrium metabolism, Progesterone metabolism, Receptors, Progesterone metabolism, Signal Transduction
- Abstract
Progesterone's ability to maintain pregnancy in eutherian mammals highlighted this steroid as the 'hormone of pregnancy'. It was the unique 'pro-gestational' bioactivity of progesterone that enabled eventual purification of this ovarian steroid to crystalline form by Willard Myron Allen in the early 1930s. While a functional connection between normal progesterone responses ('progestational proliferation') of the uterus with the maintenance of pregnancy was quickly appreciated, an understanding of progesterone's involvement in the early stages of pregnancy establishment was comparatively less well understood. With the aforementioned as historical backdrop, this review focuses on a selection of key advances in our understanding of the molecular mechanisms by which progesterone, through its nuclear receptor (the progesterone receptor), drives the development of endometrial receptivity, a transient uterine state that allows for embryo implantation and the establishment of pregnancy. Highlighted in this review are the significant contributions of advanced mouse engineering and genome-wide transcriptomic and cistromic analytics which reveal the pivotal molecular mediators and modifiers that are essential to progesterone-dependent endometrial receptivity and decidualization. With a clearer understanding of the molecular landscape that underpins uterine responsiveness to progesterone during the periimplantation period, we predict that common gynecologic morbidities due to abnormal progesterone responsiveness will be more effectively diagnosed and/or treated in the future.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.