Next Article in Journal
Special Issue “Application Progress of Liposomes in Drug Development”
Previous Article in Journal
Molecular Determinants Involved in the Docking and Uptake of Tumor-Derived Extracellular Vesicles: Implications in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Establishment of Murine Pregnancy Requires the Promyelocytic Leukemia Zinc Finger Transcription Factor

1
Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
2
Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(6), 3451; https://doi.org/10.3390/ijms25063451
Submission received: 9 February 2024 / Revised: 12 March 2024 / Accepted: 14 March 2024 / Published: 19 March 2024
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

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 Plzf d/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 Plzf d/d mice exhibit an infertility phenotype due to an inability of the embryo to implant into the Plzf d/d endometrium. Initial cellular and molecular phenotyping investigations reveal that the Plzf d/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 Plzf d/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 Plzf d/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.

1. Introduction

Although embryo abnormalities are a major cause of preclinical pregnancy loss [1,2,3,4,5], an increasing number of studies implicate the endometrium as an additional contributing factor in embryo implantation failure and early embryo miscarriage, reviewed in [6,7,8,9,10,11,12]. Given that assisted reproductive technologies (ARTs) depend on the transfer of healthy embryos into a receptive endometrium, a non-receptive endometrium at the time of embryo transfer is thought to also undercut the full potential of ART-conceived pregnancies [13,14,15,16,17]. In addition, a non-receptive endometrium is implicated as one of a number of factors leading to recurrent pregnancy loss [18,19], defined in the United States as two or more consecutive pregnancy losses that are diagnosed by ultrasound and/or histopathology [20]. Termed the adverse ripple effect [21,22], incomplete progression of the normal cellular and molecular changes in the endometrium during the peri-implantation period has also been linked with initiating adverse outcomes that symptomatically manifest in the subsequent trimesters of pregnancy; these include pre-eclampsia, placental insufficiency, intrauterine fetal restriction, and preterm birth [21].
From primate to rodent, the process of embryo implantation sequentially progresses through defined interdependent developmental stages, ostensibly beginning with blastocyst apposition and attachment to the luminal epithelium of the receptive endometrium, then embryo invasion into the subepithelial stroma, followed by decidualization of a surrounding zone of stromal fibroblasts into specialized epithelioid decidual cells; reviewed in [21,23]. Surrounding the conceptus, decidual cells support embryonic development and invasion as well as furnish protection against an adverse cytotoxic microenvironment until placentation [9].
Progesterone (P4), through the progesterone receptor (PGR), is indispensable for the majority of these early endometrial cellular changes that are required for embryo implantation, stromal decidualization, and subsequent development of the maternofetal interface [24]. Although our understanding of endometrial P4 responsiveness at the cellular level is significantly advanced, the molecular underpinnings of these cellular responses remain incomplete. A member of the evolutionary conserved POK (POZ and Kruppel) family of C2H2-type zinc finger transcription factors [25,26], the promyelocytic leukemia zinc finger (PLZF; also known as ZBTB16 or ZNF 145) can act as a transcriptional activator or repressor depending on cell and signaling context [26,27,28,29,30,31,32,33,34,35]. The PLZF transcription factor mediates a wide spectrum of early developmental programs as well as physiological effects in the adult, ranging from skeletal patterning, innate immune cell development, and hematopoiesis to spermatogenesis, reviewed in [26]. Supporting these various physiologies, PLZF controls the expansion of the stem cell pool and its differentiation, regulates cellular crosstalk, as well as governs cell proliferation, differentiation and programmed cell death, reviewed in [26].
Through an integrative approach using ChIP-seq (chromatin immunoprecipitation followed by deep sequencing) and RNA-seq datasets derived from human endometrial stromal cells (HESCs), PLZF was previously identified as a direct target of the PGR [33]. Subsequent cell-based studies demonstrated that progestin-induction of PLZF not only occurs in HESCs but was required for progestin-dependent HESC decidualization in cell culture [33,35]. This clinically significant in vitro result, along with immunohistological data showing PLZF expression occurs in human endometrial tissue, specifically during the P4-dominant secretory phase of the menstrual cycle [33], indicated that PLZF exerts an important P4 mediator role in the mammalian endometrium in vivo.
To test this proposal in vivo, a new bigenic mouse model (Plzf d/d) was generated in which PLZF function is conditionally ablated in PGR-positive cells by crossing our previously reported Pgr cre knockin mouse [36] with our recently generated Plzf floxed (Plzf f/f) mouse in which exon 2 of the Plzf gene is floxed [37]. Comparative histomorphometric, immunohistochemical, cellular, and molecular studies demonstrate that PLZF is essential for P4-dependent endometrial stromal decidualization in vivo. Furthermore, our studies on the Plzf d/d bigenic strongly support an important role for PLZF in P4-dependent endometrial receptivity, which is essential for embryo implantation and the early establishment of the maternofetal interface. Together, our mouse studies described here, along with our previously published HESC investigations [33,35], strongly support the hypothesis that the evolutionally conserved endometrial PLZF transcription factor is critical in mediating both P4-dependent uterine receptivity and decidualization.

2. Results

2.1. The PLZF Transcription Factor Is Significantly Expressed in the Murine Decidual Cell Population

To determine the expression pattern of PLZF in the endometrium of the pregnant mouse, immunohistochemical analysis was performed (Figure 1A–C). On the morning of gestation day 6 (GD6), the majority of decidual cells are immunopositive for PLZF expression within the murine decidua (Figure 1A). Immunopositivity for PLZF is located exclusively in the nucleus of decidual cells (Figure 1A), which follows a similar cellular spatial staining pattern in HESCs [33]. Outside the decidua, endometrial stromal cells within the inter-implantation sites (IIS) express significantly lower levels of PLZF, whereas a subset of glandular epithelial cells display strong PLZF expression (Figure 1B). Similar to the virgin uterus [33], PLZF expression is not detected in the murine endometrium at GD1 (Figure 1D), whereas PLZF expression is clearly evident in pre-decidual stromal fibroblasts at GD5 (Figure 1E,F). Interestingly, lower levels of PLZF are also detected in a subset of luminal and glandular epithelial cells within this tissue (Figure 1E,F). Immunohistochemical data shown in Supplementary Figure S1A–E further confirms that PLZF is expressed not only in fully differentiated decidual cells at GD6 in the mouse but also in pre-decidual stromal fibroblasts and at low levels in the luminal and glandular epithelial cellular compartments at GD5. The specificity of the PLZF antibody was confirmed using a positive control (testicular tissue derived from two-week-old male mice [38]), Supplementary Figure S1F,G.

2.2. The Plzf d/d Bigenic Uterus Is Significantly Unresponsive to Progesterone

To determine PLZF’s role in murine endometrial biology, a new bigenic mouse model was generated in which PLZF is specifically ablated in cells that express the PGR. By crossing our previously reported Pgr cre mouse [36] with a new mouse model in which exon 2 of the Plzf gene is floxed (Plzf f/f) [37], the early skeletal developmental abnormalities of the Plzf KO mouse can be avoided in the resultant conditional bigenic, termed the Plzf d/d mouse hereon. Encoding greater than 50% of the PLZF protein [39] that encompasses the initiating ATG, the N-terminal BTB/POZ domain, and the first two C2H2 zinc fingers of the PLZF DNA binding domain, the exon 2 deletion effectively abrogates PLZF expression in the Plzf KO mouse [39] and in a mouse model in which Plzf f/f is crossed with the global cytomegalovirus promoter-driven cre (CMV cre) mouse [37]. To confirm that PLZF is not expressed in the uterus of the P4-treated Plzf d/d mouse, an established P4 treatment regimen on ovariectomized mice was used (Figure 2A) [33,37].
Western immunoblot analysis clearly shows that the PLZF protein is not induced in the Plzf d/d uterus in response to acute P4 exposure (Figure 2B), a result which is also confirmed by immunohistochemistry (Figure 2C,D). While the P4-treated Plzf f/f uterus shows PLZF expression in endometrial stromal cells with low-level PLZF expression in a subset of luminal epithelial cells (Figure 2C), the Plzf d/d endometrium is devoid of PLZF immunoreactivity (Figure 2D). Because PLZF is not expressed in the P4-treated Plzf d/d endometrium (Figure 2B,D), we asked whether the expression of other P4 target genes is compromised in the absence of PLZF. Using quantitative real-time PCR (qRT-PCR), we demonstrate that the expression of the majority of the P4 responsive genes tested (Figure 2E) is either not induced or minimally induced by P4 in the Plzf d/d endometrium (Figure 2E). Together, these results confirm that the Plzf d/d mouse operates as designed and that the artificial induction of a significant number of P4 responsive genes in the ovariectomized mouse model is compromised in the Plzf d/d endometrium.

2.3. The Plzf d/d Bigenic Endometrium Fails to Support Embryo Implantation

To determine whether endometrial PLZF has a functional role during the early stages of pregnancy establishment, a standard breeding trial for at least a 6-month period was conducted using age-matched Plzf f/f and Plzf d/d females housed with fertility-proven stud males. The results clearly show that while Plzf f/f mice produce the expected number of litters with normal numbers of pups, the Plzf d/d female does not produce pups during this time period (Table 1).
The infertility phenotype displayed by the Plzf d/d female mouse occurs despite exhibiting normal ovarian activity, producing pre-implantation blastocysts similar in number to the Plzf f/f mouse, and having P4 and estradiol (E2) serum levels that are equivalent to levels detected in the Plzf f/f mouse at GD 5 (Supplementary Figure S2).
Using a timed natural pregnancy model [48], implantation sites are clearly visible following tail vein injection of Chicago Sky Blue stain along both uterine horns of the Plzf f/f mouse at GD5 (Figure 3A). In contrast, implantation sites are not detected in the Plzf d/d uterus at GD5 using the same staining procedure (Figure 3A). Follow-up immunohistochemical approaches show that while the embryo attaches to the apical surface of the luminal epithelium of the Plzf f/f endometrium at GD 5 (Figure 3B), embryo attachment does not occur in the Plzf d/d endometrium but instead floats in the open lumen (Figure 3B). Interestingly, the expression levels of a number of P4-responsive genes (Bone morphogenetic protein 2 (Bmp2) [49] and Heart and neural crest derivatives expressed 2 (Hand2) [43]), which are linked with the endometrium’s ability to support embryo implantation, are significantly attenuated in the Plzf d/d endometrium as compared with the Plzf f/f endometrium at GD 5 (Figure 3C).
Conversely, genes associated with E2-signaling (i.e., lactoferrin (Ltf) [51] and early growth response 1 (Egr1) [33,52,53,54,55]) in the endometrium are markedly elevated in the Plzf d/d endometrium as compared with the Plzf f/f endometrium (Figure 3C). These perturbations in the expression of P4 and E2 responsive genes in the Plzf d/d endometrium occur in the absence of significant changes in the expression of the estrogen receptor-α (Esr 1) and Pgr (Figure 3C). Collectively, these data support a critical role for endometrial PLZF in the early stages of embryo implantation and that the absence of PLZF derails normal E2 and P4 responsiveness of the endometrium at the molecular level.

2.4. Endometrial Receptivity Requires PLZF

Because of the impaired responsiveness of the Plzf d/d endometrium to both P4 and E2 (Figure 3C) and because sequential E2 and P4 exposure of the murine endometrium is critical for the development of the transient receptive state at GD4 [21], the cellular proliferative status of the Plzf f/f and Plzf d/d endometrium at GD4 was examined (Figure 4).
At GD4, pre-ovulatory E2-induced epithelial proliferation is significantly suppressed by rising levels of post-ovulatory P4 hormone in the normal murine endometrium [21]. As expected at GD4, the endometrium of the Plzf f/f control mouse displays a low number of epithelial cells that score positive for BrdU incorporation (Figure 4A (left panels)), whereas numerous fibroblastic cells are BrdU positive in the stromal compartment. In contrast, the Plzf d/d endometrium still retains a subset of epithelial cells that score BrdU positive as well as displaying a stromal compartment that is less proliferative than the Plzf f/f stroma at this time of gestation (Figure 4A (right panels)). These results were also confirmed using an established E2 and P4 hormone treatment protocol on ovariectomized mice that elicits an artificial receptive state [56], albeit underpinned by an exaggerated cellular proliferative response (Supplementary Figure S3). Taken together, these results reveal an unexpected role for pre-decidual PLZF in the development of the receptive endometrium that occurs prior to the development of the endometrial decidua.

2.5. Decidualization of the Murine Endometrium Is Dependent on PLZF

Because the endometrial phenotype of the Plzf d/d mouse manifests during the receptive period prior to full decidualization of the stroma, the artificial decidual response assay had to be used (Figure 5A). Using an established E2 and P4 hormone treatment regimen on ovariectomized mice that prepares the uterine horn to undergo a decidual response [57], the Plzf d/d uterus is shown to be incapable of launching a decidual response, unlike the uterus of a similarly treated Plzf f/f mouse (Figure 5A–D). At the gross morphological level, the Plzf d/d uterus fails to generate the typical deciduomata in response to the artificial deciduogenic stimulus (Figure 5A–D), in this case, sesame oil instillation into one uterine horn. The absence of a decidual response in the Plzf d/d endometrium is also accompanied by a significant decrease in the levels of established markers of murine decidualization, such as Bmp2, Hand2, Prl8a2, and Prl3c1 (Figure 5E) [43,49,50,58,59,60,61,62]. At the cellular level, there is a conspicuous absence of large epithelioid stromal decidual cells in the oil-treated Plzf d/d uterine horn, but instead, the retention of proliferative cells in the epithelium is clear (Figure 5F,H). Apart from furnishing long sought-after in vivo support for our previous human cell-based findings showing a critical role for PLZF in HESC decidualization [33,35], these results—in combination with our findings reported in Figure 4—provide strong support for PLZF’s importance not only in endometrial decidualization but also endometrial receptivity.

3. Discussion

Depending on tissue type and signaling context, PLZF has been shown to be rapidly induced by the following steroid hormones: aldosterone, cortisone, glucocorticoid, and testosterone [32,63,64,65]. These findings are significant because, like P4, these steroids exert their physiological effects through their closely related nuclear receptors of the subfamily 3 (group C) of the nuclear receptor superfamily [66]. A recent retrospective analysis of past microarray datasets revealed that support already existed as early as 2002 for the rapid induction of Plzf transcripts by P4 in the uterus of the ovariectomized mouse (Gene Expression Omnibus (GEO) profiles: Zbtb16-Progesterone effects on uterus time course (ncbi.nlm.nih.gov/geoprofiles/913691) accessed on 12 July 2023). Moreover, PLZF induction during the P4-dominant early- to mid-secretory phase of the human menstrual cycle was also previously documented in a separate microarray study (GEO profiles: ZBTB16-Endometrium throughout the menstrual cycle (ncbi.nlm.nih.gov/geoprofiles/?term=24460831) accessed on 12 July 2023). Together, these early microarray datasets provided the first indication that PLZF is an early molecular target of P4 signaling in the murine uterus and that the levels of endometrial PLZF expression are significantly elevated during the P4-dependent secretory phase of the menstrual cycle, raising the question of whether P4 induction of endometrial PLZF is essential for early pregnancy establishment. More recently, our group provided in vitro functional support for this proposal using a primary HESC culture model [33], which demonstrated that rapid progestin-induction of PLZF from basal levels in pre-decidual HESCs is essential for progestin-dependent HESC decidualization. Our subsequent in vitro studies also revealed that basal levels of PLZF in pre-decidual HESCs are essential for their proliferation, migration, and invasion [35], cellular properties known to be displayed by HESCs as they decidualize [67].
Although studies on cultured primary HESCs provided essential cellular and molecular insights into the role of PLZF in HESC decidualization, whether these in vitro functional findings translated to the in vivo situation remained an open question. Because the Plzf KO mouse exhibits a severe skeletal patterning defect [39,68], which is also observed in humans [69], we took advantage of our recently generated conditional Plzf d/d mouse that circumvents these confounding phenotypes [37]. While the initial objective in using this mouse model was to confirm our in vitro findings that PLZF is crucial for endometrial stromal cell decidualization, our first in vivo results here show that PLZF expression in pre-decidual stromal fibroblasts is critical for the development of endometrial receptivity for embryo implantation, a developmental step that precedes decidualization. Our initial molecular analysis reveals that the induction of the majority of P4-responsive molecular targets tested in this study is significantly attenuated when PLZF is absent during the peri-implantation period. Therefore, this molecular result further supports PLZF as an apex P4 mediator that is positioned high in the hierarchy of immediate P4-responsive target genes. At the cellular level, the attenuated P4 responsiveness of the Plzf d/d endometrium manifests as an incomplete suppression of E2-induced epithelial proliferation; such an unchecked heightened E2 response has been linked to implantation failure in a number of mouse models [70,71,72] and implicated in human implantation failure [73,74,75,76]. Moreover, the significant decrease in the number of proliferating pre-decidual stromal cells in the Plzf d/d endometrium at this time parallels similar findings from our previous HESC studies [33,35], where PLZF is required for P4-dependent expansion of the pre-decidual stromal cell population prior to its decidualization. The above aberrant cellular responses are underscored at the molecular level by a marked increase in the levels of E2-dependent molecular targets in Plzf d/d endometrium. In particular, the detection of elevated Egr1 levels in the Plzf d/d uterus is interesting as P4-induction of PLZF has been shown to directly suppress Egr1 expression during HESC decidualization [33]. Our past studies also demonstrate that while EGR1 expression in pre-decidual stromal fibroblasts is essential for these cells to later decidualize [35], the suppression of EGR1 expression by P4-induction of PLZF is a prerequisite if these pre-decidual stromal fibroblasts are to develop into decidual cells. As reported for HESCs [33], the levels of ESR1 and PGR are not altered in the Plzf d/d endometrium to account for these molecular changes.
Because of our past human studies [33,35], our initial objective in using the Plzf d/d mouse was to determine the in vivo importance of PLZF in murine endometrial stromal cell decidualization, which normally occurs at GD6 onwards [77]. To bypass the earlier Plzf d/d endometrial receptivity phenotype, the well-described murine artificial decidual response assay was used to determine PLZF’s endometrial intrinsic role in the decidualization of this tissue [48]. Supporting our previous human cell culture studies [33,35], the artificial decidual response assay confirmed a critical in vivo role for endometrial PLZF in the terminal differentiation of endometrial stromal fibroblasts into decidual cells. Taken together, these Plzf d/d mouse findings here underscore at least two important roles for PLZF during the peri-implantation period: (i) development of uterine receptivity to allow embryo attachment to the apical surface of the luminal epithelium and (ii) endometrial stromal fibroblast decidualization, occurring a day later in the pregnant mouse [21].
Apart from PLZF expression in pre-decidual and terminally differentiated decidual stromal cells of the murine uterus during early pregnancy, our immunohistochemical data reveal that PLZF is expressed in a subset of cells both in the luminal and glandular epithelial compartments. These observational findings indicate that PLZF may exert a previously unsuspected uterine cell-type specific role in the establishment of the maternofetal interface. With the availability of the Plzf f/f model [37], addressing the cell-type specific involvement of PLZF in uterine receptivity and decidualization is now feasible due to the recent generation of uterine cell-type specific cre mouse models. Indeed, these cre-driver models are designed not only to study the selective gene functions in the glandular and luminal epithelial compartments but also to further resolve gene functionality between two stromal cell states: the pre-decidual and decidual cell states.
While we have provided initial molecular data to support PLZF as an early mediator of P4/PGR action in the murine uterus during the peri-implantation period, genome-scale omics analysis will be essential to furnish a more comprehensive understanding of the molecular mechanisms by which PLZF exerts its physiological roles in murine uterine function. With the N-terminal BTB/POZ domain of PLZF known to be responsible for homo- and hetero-dimerization, chromatin remodeling, epigenetic transcriptional control, and formation of high-molecular-weight DNA-protein complexes [26], while PLZF’s nine C2H2 zinc finger motifs in its C-terminus are responsible for direct sequence-specific DNA binding to target genes [26], endometrial PLZF is predicted to orchestrate crucial evolutionary conserved signaling programs in response to P4. Given that we [33,35] and others [78] have shown that progestins (i.e., medroxy progesterone acetate (MPA)) induce PLZF in HESCs [33,35,78], coupled with the fact that MPA is frequently used in female reproductive medicine for the treatment of endometriosis [79], gynecological malignancies [80], postmenopausal hormone therapy [81], uterine fibroids [82], and contraceptives [83], future investigations into the molecular mechanisms that underlie this endometrial C2-H2 zinc finger transcription factor are warranted.
In summary, conclusions from these studies are that PLZF is indispensable for P4-dependent endometrial receptivity and decidualization and that PLZF may mediate these endometrial responses in a cell-type-specific manner.

4. Materials and Methods

4.1. Ethics Statement

Mice were humanely treated, and surgical procedures were conducted in accordance with the guidelines described in the Guide for the Care and Use of Laboratory Animals (“The Guide”, 8th edition, 2011), published by the National Research Council of the National Academies, Washington, DC, USA (http://www.nap.edu (accessed on 2 January 2024)). Before animal experiments were conducted, the animal protocol (AN-4203) associated with these studies was prospectively approved by the Institutional Animal Care and Use Committee at Baylor College of Medicine.

4.2. Mouse Models and Hormone Treatments

In a C57BL6J background, the Plzf conditional knockout mouse (Plzf d/d) was generated by crossing our Pgr-cre knockin (Pgr cre/+) mouse [36] with our recently generated Plzf floxed (Plzf f/f) mouse in which exon 2 of the murine Plzf gene is floxed [37]. Mice were housed in temperature-controlled rooms (22 ± 2 °C) operating on a 12 h light/12 h dark photocycle in a vivarium facility at Baylor College of Medicine, which is accredited by AAALAC (Association for Assessment and Accreditation of Laboratory Animal Care). An irradiated Formulab Diet (LabDiet/Lab Supply, Fort Worth, TX, USA (#5008)) and fresh water were made available to mice ad libitum. For acute P4 treatment, six-week-old mice were ovariectomized and rested for two weeks before receiving an intrascapular subcutaneous (s.c.) injection of P4 (1 mg) dissolved in sesame oil. Ovariectomized mice injected with sesame oil served as vehicle controls. To elicit an artificial receptive state in the uterus, eight-week-old ovariectomized mice received an E2 and P4 hormone treatment regimen as previously described [56]. Briefly, mice were initially primed with a daily injection of E2 (100 ng) for two days before resting for two days. Following the two-day rest period, mice received a daily injection of P4 (1 mg) for three days. Following the three-day P4 treatment period, mice received a combination of P4 (1 mg) and E2 (50 ng); the E2 injection at this time mimics the natural E2 nidatory spike prior to embryo implantation. Mice were euthanized 15 h following the P4E2 injection to provide uterine tissue for histological and molecular analysis. An established protocol was used to induce an artificial decidual response in the uterus of an ovariectomized mouse [57]. Briefly, eight-week-old ovariectomized mice received three daily s.c. injections of E2 (100 ng). Following two days of rest, mice were administered three daily s.c. injections of E2 (6.7 ng) plus P4 (1 mg). Six hours following the third E2P4 injection, sesame oil (50 μL) was instilled into the lumen of the left uterine horn (stimulated (S)); the right horn did not receive oil (unstimulated (U)). After intraluminal instillation of the deciduogenic stimulus, mice received daily s.c. injections of E2P4 for 5 days and then weighed before euthanasia. Trimmed of mesometrial membrane and vasculature, dissected stimulated and unstimulated uterine horns from each mouse were weighed for wet-weight measurements before further analysis.

4.3. Fertility Analyses

To time-specific days of gestation, Plzf f/f and Plzf d/d females (8–10 weeks old) were housed overnight with fertility-proven wild-type male mice. The following morning, coitus was visually confirmed in the female by the retention of a postcoital vaginal plug; the morning of detecting the vaginal plug was assigned as the morning of gestation day 1 (GD1). Prior to euthanasia on specific GDs for the described studies below, pregnant mice were individually housed. For breeding trials, sexually mature Plzf f/f control and Plzf d/d mutant females were housed with fertility-proven C57BL/6 males. Over at least a 6-month breeding period, the date of pup delivery, the number of litters, and the number of pups per litter were recorded for each female. To elicit superovulation, 21-day-old female mice were intraperitoneally (i.p.) injected with pregnant mare serum gonadotropin (PMSG; Sigma-Aldrich, St. Louis, MO, USA (5 international units (IU)/100 μL of sterile 0.9% saline)). Forty-eight hours post-injection, mice received an i.p. injection of human chorionic gonadotropin (hCG; Sigma-Aldrich (5 IU/100 μL of sterile 0.9% saline)). Sixteen hours later, oocytes were harvested from the lumen of the fallopian tubes and counted using a dissecting microscope as previously detailed [84]. Using fallopian tube tissue that was dissected from Plzf f/f and Plzf d/d females at GD 2, two-cell stage embryos were retrieved by gently flushing the fallopian tube lumen with 100 μL of sterile phosphate-buffered saline (PBS). To visualize emerging implantation sites along both uterine horns of mice at GD5, a Chicago sky blue dye solution (1% in PBS; 100 μL per mouse) was injected into one lateral tail vein before mice were euthanized 2–5 min later [84]; stained reproductive tract tissues were removed first for the purposes of imaging and subsequent cellular and molecular analyses.

4.4. Histology and Immunohistochemical Detection

Following overnight fixation in 4% paraformaldehyde, tissues were stepwise dehydrated by sequential incubation in increased concentrations of ethanol; dehydrated and fixed tissues were cleared with xylene before paraffin embedding. Paraffin-embedded tissues were sectioned to 5 μm thickness onto slides before tissue sections were stained with hematoxylin and eosin (H&E) for general histological analysis [84,85]. Immunohistochemical visualization of PLZF protein was achieved using an anti-PLZF primary mouse monoclonal antibody (D-9 [86]; Santa Cruz Biotechnology Inc., Dallas, TX, USA, #sc-28319, 1:150 dilution) in combination with the Mouse on Mouse (M.O.M.) Elite Immunodetection kit, Peroxidase (Vector Laboratories Inc., Burlingame, CA, PK-2200). Using the BrdU in situ detection kit (BD Biosciences, San Jose CA, USA, #551321), cells in the S-phase of the cell cycle were visualized by immunohistochemical detection of 5-bromo-2′-deoxyuridine (BrdU). After immunostaining, tissue sections were counterstained with hematoxylin prior to the application of Permount solution to aid the placement of coverslips. For cell counting in general, immunopositive cells were counted within a field of 300 cells to obtain the mean number of immunopositive cells per 300 cells counted. For the majority of studies, at least 3–4 uterine fields of 300 cells were counted per mouse. Unless otherwise specified, the mean number of immunopositive cells was calculated from at least 3 mice per genotype and treatment group. Recent versions of the Photoshop and Illustrator programs within the Adobe Creative Suite software package (Adobe Systems Inc. San Jose, CA, USA (https://www.adobe.com/creativecloud/plans.html)) were used for raw image processing, compiling of image composites, and the annotation of the final figures for manuscript preparation.

4.5. Molecular Analysis

Experimental conditions for murine PLZF immunoblot analyses have been reported previously [33,37]. Briefly, protein concentration was obtained using the Bradford reagent (ThermoFisher Scientific Inc., Waltham, MA, USA, #23225) before protein extracts (20 μg/lane) were resolved on a 4–15% polyacrylamide-SDS gel. Resolved proteins were transferred to a polyvinylidene difluoride (PVDF) membrane. After blocking PVDF membranes with 5% nonfat milk in Tris-buffered saline containing 0.1% Tween 20 (TBST), membranes were incubated with an anti-PLZF primary mouse monoclonal antibody (D-9; Santa Cruz Biotechnology Inc. # sc-28319) or a mouse monoclonal anti-β-actin (AC-74, Sigma-Aldrich; #A2228) overnight at 4 °C. Following the primary antibody incubation step, the immunoblots were washed and then incubated with an anti-mouse IgG1k horseradish peroxidase (HRP)-conjugated secondary antibody (m-IgG Fc BP-HRP; Santa Cruz Biotechnology Inc., sc-525409) for 1 h at room temperature. Resultant chemiluminescence signals were detected using the SuperSignal West Pico PLUS Chemiluminescent substrate kit (ThermoFisher Scientific Inc., #34580). For quantitative real-time PCR (qRT-PCR) analyses, total RNA was prepared from uterine tissue using the RNeasy Plus Mini kit (Qiagen Inc., Germantown, MD #74134). The NanoDrop 2000 UV/Visual spectrophotometer (ThermoFisher Scientific Inc.) was used to quantitate RNA before the reverse transcription step using the High-Capacity cDNA Reverse Transcription kit (ThermoFisher Scientific Inc. #4368814). Following reverse transcription, amplified cDNA was diluted to 10 ng/μL before qRT-PCR was conducted using the Fast TaqMan 2X Mastermix (Applied Biosystems/Life Technologies, Grand Island, NY, USA, #4352042); the TaqMan assays used in these experiments are listed in Table S1. All qRT-PCR experiments were performed on the 7500 Fast Real-Time PCR system (Applied Biosystems/Life Technologies); the delta-delta cycle threshold was used to normalize expression to the internal 18S reference.

4.6. Serum Hormone Measurements

Using microtainer tubes containing a serum separator microguard (Becton, Dickinson and Company, Franklin Lakes, NJ, USA (#365967)), whole blood was collected from virgin and pregnant mice at GD5 as previously described [85]. To ensure complete blood coagulation, drawn blood was held at room temperature for 30 min before serum samples were separated by centrifugation at 2000 rpm for 10 min at 4 °C; serum was stored at −80 °C until analysis. Serum P4 and E2 levels were measured by the Ligand Assay and Analysis Core of the Center for Research in Reproduction at the University of Virginia (Charlottesville, VA, USA); assay details are available at https://med.virginia.edu/research-in-reproduction/ligand-assay-analysis-core/assay-methods/ (accessed on 15 October 2023).

4.7. Statistical Analyses

The two-tailed unpaired Student t-test (Welch-corrected) was used to estimate the statistical significance of differences between the two groups. Unless otherwise stated, data were graphically displayed as the mean ± standard error of the mean (s.e.m.). Differences between means with a p-value < 0.05 were considered statistically significant, with the number of asterisks indicating the level of significance: * p < 0.05; ** p < 0.01; and *** p < 0.001. Version 9 of the Prism software package from GraphPad Software Inc. (San Diego, CA, USA) was used for the statistical analyses in these studies.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms25063451/s1.

Author Contributions

L.H., V.K.M., F.J.D. and J.P.L. developed the experimental mouse models and designed the research experiments. All authors contributed to the drafting, editing, and reviewing of the manuscript prior to submission. L.H. and V.K.M. conducted the experiments as well as acquired and analyzed the data, which were reviewed by F.J.D. and J.P.L. L.H. generated the Plzf d/d bigenic mouse model and conducted the initial phenotype experiments and V.K.M. performed the required histological and molecular analysis of the reproductive phenotype. All authors have read and agreed to the published version of the manuscript.

Funding

This research study was funded in part by the National Institute of Environmental Health Sciences (NIEHS) Grant/Award Number: Z1AE8103311-01 to F.J.D. and by the National Institutes of Health (NIH)/National Institute of Child Health and Human Development (NICHD) Grant/Award Number: R01 HD-042311 to J.P.L.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Animal Care and Use Committee (IACUC) of Baylor College of Medicine (AN-4203; approved 10/24/2023).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data supporting the findings of this study are available within the article and Supplementary Materials.

Acknowledgments

We like to thank Yan Ying and Rong Zhao for their essential technical expertise.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.

References

  1. Boue, J.; Bou, A.; Lazar, P. Retrospective and prospective epidemiological studies of 1500 karyotyped spontaneous human abortions. Teratology 1975, 12, 11–26. [Google Scholar] [CrossRef]
  2. Eiben, B.; Borgmann, S.; Schubbe, I.; Hansmann, I. A cytogenetic study directly from chorionic villi of 140 spontaneous abortions. Hum. Genet. 1987, 77, 137–141. [Google Scholar] [CrossRef]
  3. Farfalli, V.I.; Magli, M.C.; Ferraretti, A.P.; Gianaroli, L. Role of aneuploidy on embryo implantation. Gynecol. Obstet. Investig. 2007, 64, 161–165. [Google Scholar] [CrossRef]
  4. Findikli, N.; Kahraman, S.; Saglam, Y.; Beyazyurek, C.; Sertyel, S.; Karlikaya, G.; Karagozoglu, H.; Aygun, B. Embryo aneuploidy screening for repeated implantation failure and unexplained recurrent miscarriage. Reprod. Biomed. Online 2006, 13, 38–46. [Google Scholar] [CrossRef] [PubMed]
  5. Quenby, S.; Vince, G.; Farquharson, R.; Aplin, J. Recurrent miscarriage: A defect in nature’s quality control? Hum. Reprod. 2002, 17, 1959–1963. [Google Scholar] [CrossRef] [PubMed]
  6. Bellver, J.; Simon, C. Implantation failure of endometrial origin: What is new? Curr. Opin. Obstet. Gynecol. 2018, 30, 229–236. [Google Scholar] [CrossRef]
  7. Bui, A.H.; Timmons, D.B.; Young, S.L. Evaluation of endometrial receptivity and implantation failure. Curr. Opin. Obstet. Gynecol. 2022, 34, 107–113. [Google Scholar] [CrossRef]
  8. Macklon, N.S.; Geraedts, J.P.; Fauser, B.C. Conception to ongoing pregnancy: The ‘black box’ of early pregnancy loss. Hum. Reprod. Update 2002, 8, 333–343. [Google Scholar] [CrossRef]
  9. Muter, J.; Lynch, V.J.; McCoy, R.C.; Brosens, J.J. Human embryo implantation. Development 2023, 150, dev201507. [Google Scholar] [CrossRef] [PubMed]
  10. Ng, S.W.; Norwitz, G.A.; Pavlicev, M.; Tilburgs, T.; Simon, C.; Norwitz, E.R. Endometrial Decidualization: The Primary Driver of Pregnancy Health. Int. J. Mol. Sci. 2020, 21, 4092. [Google Scholar] [CrossRef]
  11. Pathare, A.D.S.; Loid, M.; Saare, M.; Gidlof, S.B.; Zamani Esteki, M.; Acharya, G.; Peters, M.; Salumets, A. Endometrial receptivity in women of advanced age: An underrated factor in infertility. Hum. Reprod. Update 2023, 29, 773–793. [Google Scholar] [CrossRef]
  12. Ticconi, C.; Di Simone, N.; Campagnolo, L.; Fazleabas, A. Clinical consequences of defective decidualization. Tissue Cell 2021, 72, 101586. [Google Scholar] [CrossRef]
  13. Galliano, D.; Bellver, J.; Diaz-Garcia, C.; Simon, C.; Pellicer, A. ART and uterine pathology: How relevant is the maternal side for implantation? Hum. Reprod. Update 2015, 21, 13–38. [Google Scholar] [CrossRef]
  14. Savasi, V.; Leone, F.P.; Fuse, F.; Parisi, F.; Cetin, I. Assisted reproductive technologies and uterine factors influencing their success. Minerva Ginecol. 2013, 65, 505–524. [Google Scholar] [PubMed]
  15. Fatemi, H.M.; Popovic-Todorovic, B. Implantation in assisted reproduction: A look at endometrial receptivity. Reprod. Biomed. Online 2013, 27, 530–538. [Google Scholar] [CrossRef]
  16. Simon, C. Endometrial factor testing is a useful tool in clinical infertility management. Reprod. Biomed. Online 2022, 44, 953–960. [Google Scholar] [CrossRef] [PubMed]
  17. Coulam, C.B.; Chapman, C.; Rinehart, J.S. What is a preclinical pregnancy loss? J. Assist. Reprod. Genet. 1998, 15, 184–187. [Google Scholar] [CrossRef]
  18. Santamaria, X.; Simon, C. Endometrial Factor in Unexplained Infertility and Recurrent Implantation Failure. Semin. Reprod. Med. 2021, 39, 227–232. [Google Scholar] [CrossRef] [PubMed]
  19. Ma, J.; Gao, W.; Li, D. Recurrent implantation failure: A comprehensive summary from etiology to treatment. Front. Endocrinol. 2022, 13, 1061766. [Google Scholar] [CrossRef]
  20. Practice Committee of the American Society for Reproductive, M. Definitions of infertility and recurrent pregnancy loss: A committee opinion. Fertil. Steril. 2013, 99, 63. [Google Scholar] [CrossRef]
  21. Cha, J.; Sun, X.; Dey, S.K. Mechanisms of implantation: Strategies for successful pregnancy. Nat. Med. 2012, 18, 1754–1767. [Google Scholar] [CrossRef]
  22. Kelleher, A.M.; Milano-Foster, J.; Behura, S.K.; Spencer, T.E. Uterine glands coordinate on-time embryo implantation and impact endometrial decidualization for pregnancy success. Nat. Commun. 2018, 9, 2435. [Google Scholar] [CrossRef]
  23. Gellersen, B.; Brosens, J.J. Cyclic decidualization of the human endometrium in reproductive health and failure. Endocr. Rev. 2014, 35, 851–905. [Google Scholar] [CrossRef]
  24. Cope, D.I.; Monsivais, D. Progesterone Receptor Signaling in the Uterus Is Essential for Pregnancy Success. Cells 2022, 11, 1474. [Google Scholar] [CrossRef]
  25. Liu, T.M.; Lee, E.H.; Lim, B.; Shyh-Chang, N. Concise Review: Balancing Stem Cell Self-Renewal and Differentiation with PLZF. Stem Cells 2016, 34, 277–287. [Google Scholar] [CrossRef]
  26. Suliman, B.A.; Xu, D.; Williams, B.R. The promyelocytic leukemia zinc finger protein: Two decades of molecular oncology. Front. Oncol. 2012, 2, 74. [Google Scholar] [CrossRef]
  27. Mao, A.P.; Constantinides, M.G.; Mathew, R.; Zuo, Z.; Chen, X.; Weirauch, M.T.; Bendelac, A. Multiple layers of transcriptional regulation by PLZF in NKT-cell development. Proc. Natl. Acad. Sci. USA 2016, 113, 7602–7607. [Google Scholar] [CrossRef]
  28. Lovelace, D.L.; Gao, Z.; Mutoji, K.; Song, Y.C.; Ruan, J.; Hermann, B.P. The regulatory repertoire of PLZF and SALL4 in undifferentiated spermatogonia. Development 2016, 143, 1893–1906. [Google Scholar] [CrossRef]
  29. Labbaye, C.; Quaranta, M.T.; Pagliuca, A.; Militi, S.; Licht, J.D.; Testa, U.; Peschle, C. PLZF induces megakaryocytic development, activates Tpo receptor expression and interacts with GATA1 protein. Oncogene 2002, 21, 6669–6679. [Google Scholar] [CrossRef]
  30. Ikeda, R.; Yoshida, K.; Tsukahara, S.; Sakamoto, Y.; Tanaka, H.; Furukawa, K.; Inoue, I. The promyelotic leukemia zinc finger promotes osteoblastic differentiation of human mesenchymal stem cells as an upstream regulator of CBFA1. J. Biol. Chem. 2005, 280, 8523–8530. [Google Scholar] [CrossRef]
  31. Gaber, Z.B.; Butler, S.J.; Novitch, B.G. PLZF regulates fibroblast growth factor responsiveness and maintenance of neural progenitors. PLoS Biol. 2013, 11, e1001676. [Google Scholar] [CrossRef]
  32. Fahnenstich, J.; Nandy, A.; Milde-Langosch, K.; Schneider-Merck, T.; Walther, N.; Gellersen, B. Promyelocytic leukaemia zinc finger protein (PLZF) is a glucocorticoid- and progesterone-induced transcription factor in human endometrial stromal cells and myometrial smooth muscle cells. Mol. Hum. Reprod. 2003, 9, 611–623. [Google Scholar] [CrossRef]
  33. Kommagani, R.; Szwarc, M.M.; Vasquez, Y.M.; Peavey, M.C.; Mazur, E.C.; Gibbons, W.E.; Lanz, R.B.; DeMayo, F.J.; Lydon, J.P. The Promyelocytic Leukemia Zinc Finger Transcription Factor Is Critical for Human Endometrial Stromal Cell Decidualization. PLoS Genet. 2016, 12, e1005937. [Google Scholar] [CrossRef]
  34. Singh, S.P.; Zhang, H.H.; Tsang, H.; Gardina, P.J.; Myers, T.G.; Nagarajan, V.; Lee, C.H.; Farber, J.M. PLZF regulates CCR6 and is critical for the acquisition and maintenance of the Th17 phenotype in human cells. J. Immunol. 2015, 194, 4350–4361. [Google Scholar] [CrossRef]
  35. Szwarc, M.M.; Hai, L.; Gibbons, W.E.; Peavey, M.C.; White, L.D.; Mo, Q.; Lonard, D.M.; Kommagani, R.; Lanz, R.B.; DeMayo, F.J.; et al. Human endometrial stromal cell decidualization requires transcriptional reprogramming by PLZF. Biol. Reprod. 2018, 98, 15–27. [Google Scholar] [CrossRef]
  36. Soyal, S.M.; Mukherjee, A.; Lee, K.Y.; Li, J.; Li, H.; DeMayo, F.J.; Lydon, J.P. Cre-mediated recombination in cell lineages that express the progesterone receptor. Genesis 2005, 41, 58–66. [Google Scholar] [CrossRef]
  37. Hai, L.; Szwarc, M.M.; Lanza, D.G.; Heaney, J.D.; Lydon, J.P. Using CRISPR/Cas9 engineering to generate a mouse with a conditional knockout allele for the promyelocytic leukemia zinc finger transcription factor. Genesis 2019, 57, e23281. [Google Scholar] [CrossRef]
  38. Costoya, J.A.; Hobbs, R.M.; Barna, M.; Cattoretti, G.; Manova, K.; Sukhwani, M.; Orwig, K.E.; Wolgemuth, D.J.; Pandolfi, P.P. Essential role of Plzf in maintenance of spermatogonial stem cells. Nat. Genet. 2004, 36, 653–659. [Google Scholar] [CrossRef]
  39. Barna, M.; Hawe, N.; Niswander, L.; Pandolfi, P.P. Plzf regulates limb and axial skeletal patterning. Nat. Genet. 2000, 25, 166–172. [Google Scholar] [CrossRef]
  40. Das, S.K.; Chakraborty, I.; Paria, B.C.; Wang, X.N.; Plowman, G.; Dey, S.K. Amphiregulin is an implantation-specific and progesterone-regulated gene in the mouse uterus. Mol. Endocrinol. 1995, 9, 691–705. [Google Scholar]
  41. Han, B.C.; Xia, H.F.; Sun, J.; Yang, Y.; Peng, J.P. Retinoic acid-metabolizing enzyme cytochrome P450 26a1 (cyp26a1) is essential for implantation: Functional study of its role in early pregnancy. J. Cell Physiol. 2010, 223, 471–479. [Google Scholar] [CrossRef]
  42. Kim, T.H.; Lee, D.K.; Franco, H.L.; Lydon, J.P.; Jeong, J.W. ERBB receptor feedback inhibitor 1 regulation of estrogen receptor activity is critical for uterine implantation in mice. Biol. Reprod. 2010, 82, 706–713. [Google Scholar] [CrossRef] [PubMed]
  43. Li, Q.; Kannan, A.; DeMayo, F.J.; Lydon, J.P.; Cooke, P.S.; Yamagishi, H.; Srivastava, D.; Bagchi, M.K.; Bagchi, I.C. The antiproliferative action of progesterone in uterine epithelium is mediated by Hand2. Science 2011, 331, 912–916. [Google Scholar] [CrossRef]
  44. Paria, B.C.; Das, N.; Das, S.K.; Zhao, X.; Dileepan, K.N.; Dey, S.K. Histidine decarboxylase gene in the mouse uterus is regulated by progesterone and correlates with uterine differentiation for blastocyst implantation. Endocrinology 1998, 139, 3958–3966. [Google Scholar] [CrossRef]
  45. Lee, K.; Jeong, J.; Kwak, I.; Yu, C.T.; Lanske, B.; Soegiarto, D.W.; Toftgard, R.; Tsai, M.J.; Tsai, S.; Lydon, J.P.; et al. Indian hedgehog is a major mediator of progesterone signaling in the mouse uterus. Nat. Genet. 2006, 38, 1204–1209. [Google Scholar] [CrossRef]
  46. Marquardt, R.M.; Lee, K.; Kim, T.H.; Lee, B.; DeMayo, F.J.; Jeong, J.W. Interleukin-13 receptor subunit alpha-2 is a target of progesterone receptor and steroid receptor coactivator-1 in the mouse uterusdagger. Biol. Reprod. 2020, 103, 760–768. [Google Scholar] [CrossRef]
  47. Kommagani, R.; Szwarc, M.M.; Kovanci, E.; Creighton, C.J.; O’Malley, B.W.; Demayo, F.J.; Lydon, J.P. A murine uterine transcriptome, responsive to steroid receptor coactivator-2, reveals transcription factor 23 as essential for decidualization of human endometrial stromal cells. Biol. Reprod. 2014, 90, 75. [Google Scholar] [CrossRef]
  48. Deb, K.; Reese, J.; Paria, B.C. Methodologies to study implantation in mice. Methods Mol. Med. 2006, 121, 9–34. [Google Scholar]
  49. Lee, K.Y.; Jeong, J.W.; Wang, J.; Ma, L.; Martin, J.F.; Tsai, S.Y.; Lydon, J.P.; DeMayo, F.J. Bmp2 is critical for the murine uterine decidual response. Mol. Cell Biol. 2007, 27, 5468–5478. [Google Scholar] [CrossRef]
  50. Li, Q.; Kannan, A.; Wang, W.; Demayo, F.J.; Taylor, R.N.; Bagchi, M.K.; Bagchi, I.C. Bone morphogenetic protein 2 functions via a conserved signaling pathway involving Wnt4 to regulate uterine decidualization in the mouse and the human. J. Biol. Chem. 2007, 282, 31725–31732. [Google Scholar] [CrossRef]
  51. Pentecost, B.T.; Teng, C.T. Lactotransferrin is the major estrogen inducible protein of mouse uterine secretions. J. Biol. Chem. 1987, 262, 10134–10139. [Google Scholar] [CrossRef]
  52. Cicatiello, L.; Sica, V.; Bresciani, F.; Weisz, A. Identification of a specific pattern of “immediate-early” gene activation induced by estrogen during mitogenic stimulation of rat uterine cells. Receptor 1993, 3, 17–30. [Google Scholar]
  53. Guo, B.; Tian, X.C.; Li, D.D.; Yang, Z.Q.; Cao, H.; Zhang, Q.L.; Liu, J.X.; Yue, Z.P. Expression, regulation and function of Egr1 during implantation and decidualization in mice. Cell Cycle 2014, 13, 2626–2640. [Google Scholar] [CrossRef]
  54. Kim, H.R.; Kim, Y.S.; Yoon, J.A.; Yang, S.C.; Park, M.; Seol, D.W.; Lyu, S.W.; Jun, J.H.; Lim, H.J.; Lee, D.R.; et al. Estrogen induces EGR1 to fine-tune its actions on uterine epithelium by controlling PR signaling for successful embryo implantation. FASEB J. 2018, 32, 1184–1195. [Google Scholar] [CrossRef]
  55. Park, M.; Kim, H.R.; Kim, Y.S.; Yang, S.C.; Yoon, J.A.; Lyu, S.W.; Lim, H.J.; Hong, S.H.; Song, H. Estrogen-induced transcription factor EGR1 regulates c-Kit transcription in the mouse uterus to maintain uterine receptivity for embryo implantation. Mol. Cell Endocrinol. 2018, 470, 75–83. [Google Scholar] [CrossRef]
  56. Tong, W.; Pollard, J.W. Progesterone inhibits estrogen-induced cyclin D1 and cdk4 nuclear translocation, cyclin E- and cyclin A-cdk2 kinase activation, and cell proliferation in uterine epithelial cells in mice. Mol. Cell Biol. 1999, 19, 2251–2264. [Google Scholar] [CrossRef]
  57. Zhang, Y.; Zhang, Z.; Kang, N.; Sheng, X. Generation of a Mouse Artificial Decidualization Model with Ovariectomy for Endometrial Decidualization Research. J. Vis. Exp. 2022, 185, e64278. [Google Scholar] [CrossRef]
  58. Huyen, D.V.; Bany, B.M. Evidence for a conserved function of heart and neural crest derivatives expressed transcript 2 in mouse and human decidualization. Reproduction 2011, 142, 353–368. [Google Scholar] [CrossRef]
  59. Shindoh, H.; Okada, H.; Tsuzuki, T.; Nishigaki, A.; Kanzaki, H. Requirement of heart and neural crest derivatives-expressed transcript 2 during decidualization of human endometrial stromal cells in vitro. Fertil. Steril. 2014, 101, 1781–1790.e1-5. [Google Scholar] [CrossRef]
  60. Li, Q.; Kannan, A.; Das, A.; Demayo, F.J.; Hornsby, P.J.; Young, S.L.; Taylor, R.N.; Bagchi, M.K.; Bagchi, I.C. WNT4 acts downstream of BMP2 and functions via beta-catenin signaling pathway to regulate human endometrial stromal cell differentiation. Endocrinology 2013, 154, 446–457. [Google Scholar] [CrossRef]
  61. Alam, S.M.; Konno, T.; Sahgal, N.; Lu, L.; Soares, M.J. Decidual cells produce a heparin-binding prolactin family cytokine with putative intrauterine regulatory actions. J. Biol. Chem. 2008, 283, 18957–18968. [Google Scholar] [CrossRef]
  62. Alam, S.M.; Konno, T.; Soares, M.J. Identification of target genes for a prolactin family paralog in mouse decidua. Reproduction 2015, 149, 625–632. [Google Scholar] [CrossRef]
  63. Naray-Fejes-Toth, A.; Boyd, C.; Fejes-Toth, G. Regulation of epithelial sodium transport by promyelocytic leukemia zinc finger protein. Am. J. Physiol. Renal Physiol. 2008, 295, F18–F26. [Google Scholar] [CrossRef]
  64. Jiang, F.; Wang, Z. Identification and characterization of PLZF as a prostatic androgen-responsive gene. Prostate 2004, 59, 426–435. [Google Scholar] [CrossRef]
  65. Peppi, M.; Kujawa, S.G.; Sewell, W.F. A corticosteroid-responsive transcription factor, promyelocytic leukemia zinc finger protein, mediates protection of the cochlea from acoustic trauma. J. Neurosci. 2011, 31, 735–741. [Google Scholar] [CrossRef]
  66. Mangelsdorf, D.J.; Thummel, C.; Beato, M.; Herrlich, P.; Schutz, G.; Umesono, K.; Blumberg, B.; Kastner, P.; Mark, M.; Chambon, P.; et al. The nuclear receptor superfamily: The second decade. Cell 1995, 83, 835–839. [Google Scholar] [CrossRef]
  67. Weimar, C.H.; Macklon, N.S.; Post Uiterweer, E.D.; Brosens, J.J.; Gellersen, B. The motile and invasive capacity of human endometrial stromal cells: Implications for normal and impaired reproductive function. Hum. Reprod. Update 2013, 19, 542–557. [Google Scholar] [CrossRef]
  68. Barna, M.; Pandolfi, P.P.; Niswander, L. Gli3 and Plzf cooperate in proximal limb patterning at early stages of limb development. Nature 2005, 436, 277–281. [Google Scholar] [CrossRef]
  69. Fischer, S.; Kohlhase, J.; Bohm, D.; Schweiger, B.; Hoffmann, D.; Heitmann, M.; Horsthemke, B.; Wieczorek, D. Biallelic loss of function of the promyelocytic leukaemia zinc finger (PLZF) gene causes severe skeletal defects and genital hypoplasia. J. Med. Genet. 2008, 45, 731–737. [Google Scholar] [CrossRef]
  70. Kurihara, I.; Lee, D.K.; Petit, F.G.; Jeong, J.; Lee, K.; Lydon, J.P.; DeMayo, F.J.; Tsai, M.J.; Tsai, S.Y. COUP-TFII mediates progesterone regulation of uterine implantation by controlling ER activity. PLoS Genet. 2007, 3, e102. [Google Scholar] [CrossRef]
  71. Ma, W.G.; Song, H.; Das, S.K.; Paria, B.C.; Dey, S.K. Estrogen is a critical determinant that specifies the duration of the window of uterine receptivity for implantation. Proc. Natl. Acad. Sci. USA 2003, 100, 2963–2968. [Google Scholar] [CrossRef]
  72. Sun, X.; Bartos, A.; Whitsett, J.A.; Dey, S.K. Uterine deletion of Gp130 or Stat3 shows implantation failure with increased estrogenic responses. Mol. Endocrinol. 2013, 27, 1492–1501. [Google Scholar] [CrossRef]
  73. Horcajadas, J.A.; Diaz-Gimeno, P.; Pellicer, A.; Simon, C. Uterine receptivity and the ramifications of ovarian stimulation on endometrial function. Semin. Reprod. Med. 2007, 25, 454–460. [Google Scholar] [CrossRef]
  74. Loutradis, D.; Beretsos, P.; Arabatzi, E.; Anagnostou, E.; Drakakis, P. The role of steroid hormones in ART. J. Steroid Biochem. Mol. Biol. 2008, 112, 1–4. [Google Scholar] [CrossRef]
  75. Pellicer, A.; Valbuena, D.; Cano, F.; Remohi, J.; Simon, C. Lower implantation rates in high responders: Evidence for an altered endocrine milieu during the preimplantation period. Fertil. Steril. 1996, 65, 1190–1195. [Google Scholar] [CrossRef]
  76. Simon, C.; Garcia Velasco, J.J.; Valbuena, D.; Peinado, J.A.; Moreno, C.; Remohi, J.; Pellicer, A. Increasing uterine receptivity by decreasing estradiol levels during the preimplantation period in high responders with the use of a follicle-stimulating hormone step-down regimen. Fertil. Steril. 1998, 70, 234–239. [Google Scholar] [CrossRef]
  77. Wang, H.; Dey, S.K. Roadmap to embryo implantation: Clues from mouse models. Nat. Rev. Genet. 2006, 7, 185–199. [Google Scholar] [CrossRef]
  78. Arlier, S.; Kayisli, U.A.; Semerci, N.; Ozmen, A.; Larsen, K.; Schatz, F.; Lockwood, C.J.; Guzeloglu-Kayisli, O. Enhanced ZBTB16 Levels by Progestin-Only Contraceptives Induces Decidualization and Inflammation. Int. J. Mol. Sci. 2023, 24, 10532. [Google Scholar] [CrossRef]
  79. Clemenza, S.; Capezzuoli, T.; Eren, E.; Garcia Garcia, J.M.; Vannuccini, S.; Petraglia, F. Progesterone receptor ligands for the treatment of endometriosis. Minerva Obstet. Gynecol. 2023, 75, 288–297. [Google Scholar] [CrossRef]
  80. Ramirez, P.T.; Frumovitz, M.; Bodurka, D.C.; Sun, C.C.; Levenback, C. Hormonal therapy for the management of grade 1 endometrial adenocarcinoma: A literature review. Gynecol. Oncol. 2004, 95, 133–138. [Google Scholar] [CrossRef]
  81. Stanczyk, F.Z.; Bhavnani, B.R. Use of medroxyprogesterone acetate for hormone therapy in postmenopausal women: Is it safe? J. Steroid Biochem. Mol. Biol. 2014, 142, 30–38. [Google Scholar] [CrossRef]
  82. Moro, E.; Degli Esposti, E.; Borghese, G.; Manzara, F.; Zanello, M.; Raimondo, D.; Gava, G.; Arena, A.; Casadio, P.; Meriggiola, M.C.; et al. The Impact of Hormonal Replacement Treatment in Postmenopausal Women with Uterine Fibroids: A State-of-the-Art Review of the Literature. Medicina 2019, 55, 549. [Google Scholar] [CrossRef]
  83. Westhoff, C. Depot-medroxyprogesterone acetate injection (Depo-Provera): A highly effective contraceptive option with proven long-term safety. Contraception 2003, 68, 75–87. [Google Scholar] [CrossRef]
  84. Maurya, V.K.; Szwarc, M.M.; Lonard, D.M.; Kommagani, R.; Wu, S.P.; O’Malley, B.W.; DeMayo, F.J.; Lydon, J.P. Steroid receptor coactivator-2 drives epithelial reprogramming that enables murine embryo implantation. FASEB J. 2023, 37, e23313. [Google Scholar] [CrossRef]
  85. Hai, L.; Szwarc, M.M.; He, B.; Lonard, D.M.; Kommagani, R.; DeMayo, F.J.; Lydon, J.P. Uterine function in the mouse requires speckle-type poz protein. Biol. Reprod. 2018, 98, 856–869. [Google Scholar] [CrossRef]
  86. Hu, H.; Sun, N.; Du, H.; He, Y.; Pan, K.; Liu, X.; Lu, X.; Wei, J.; Liao, M.; Duan, C. Mouse promyelocytic leukemia zinc finger protein (PLZF) regulates hepatic lipid and glucose homeostasis dependent on SIRT1. Front. Pharmacol. 2022, 13, 1039726. [Google Scholar] [CrossRef]
Figure 1. The murine decidua expresses the PLZF transcription factor. (A) Immunohistochemistry detects PLZF expression in the majority of decidual cells in the decidual bed of the mouse at GD 6 (black arrowhead); white arrowhead indicates a small subset of decidual cells that are PLZF negative. (B) Immunohistochemistry also reveals PLZF expression in the majority of glandular epithelial (GE) cells (black arrowhead), whereas the majority of stromal fibroblasts in the inter-implantation site (IIS) region express low levels of PLZF (white arrowhead); the vertical dashed line indicates the border between the periphery of the decidua and the IIS region. The scale bar in (A) applies to (B). (C) A longitudinal section of the uterine horn with five decidual beds (numbered 1–5), which is immunohistochemically stained for PLZF expression, is shown. The image in (C) was captured at low magnification of one uterine horn from which the higher magnification immunohistochemical data shown in panels (A,B) are derived. The antimesometrial and mesometrial poles are indicated by AM and M, respectively; one of the IIS regions is indicated by a bracket. (D) There is an absence of obvious PLZF immunopositivity in the murine uterus on the afternoon of GD 1 (white arrowheads); LE and S denote luminal epithelium and stroma, respectively. (E) On the morning of GD5, low immunopositivity for PLZF can be detected in a subset of luminal epithelial cells (grey arrowhead), while the remaining luminal epithelial cell population scores negative for PLZF expression (white arrowhead). The glandular epithelium (GE) exhibits a similar PLZF pattern as described for the luminal epithelium. The black arrowhead indicates a PLZF positive stromal cell. (F) The majority of the cells in the stromal (S) compartment of the GD5 endometrium are strongly immunopositive for PLZF expression (black arrowhead), whereas only a small subset of stromal cells is PLZF negative (white arrowhead). The grey arrowhead indicates a glandular epithelial cell that is positive for PLZF expression. The scale bar in (D) applies to both panels (E,F).
Figure 1. The murine decidua expresses the PLZF transcription factor. (A) Immunohistochemistry detects PLZF expression in the majority of decidual cells in the decidual bed of the mouse at GD 6 (black arrowhead); white arrowhead indicates a small subset of decidual cells that are PLZF negative. (B) Immunohistochemistry also reveals PLZF expression in the majority of glandular epithelial (GE) cells (black arrowhead), whereas the majority of stromal fibroblasts in the inter-implantation site (IIS) region express low levels of PLZF (white arrowhead); the vertical dashed line indicates the border between the periphery of the decidua and the IIS region. The scale bar in (A) applies to (B). (C) A longitudinal section of the uterine horn with five decidual beds (numbered 1–5), which is immunohistochemically stained for PLZF expression, is shown. The image in (C) was captured at low magnification of one uterine horn from which the higher magnification immunohistochemical data shown in panels (A,B) are derived. The antimesometrial and mesometrial poles are indicated by AM and M, respectively; one of the IIS regions is indicated by a bracket. (D) There is an absence of obvious PLZF immunopositivity in the murine uterus on the afternoon of GD 1 (white arrowheads); LE and S denote luminal epithelium and stroma, respectively. (E) On the morning of GD5, low immunopositivity for PLZF can be detected in a subset of luminal epithelial cells (grey arrowhead), while the remaining luminal epithelial cell population scores negative for PLZF expression (white arrowhead). The glandular epithelium (GE) exhibits a similar PLZF pattern as described for the luminal epithelium. The black arrowhead indicates a PLZF positive stromal cell. (F) The majority of the cells in the stromal (S) compartment of the GD5 endometrium are strongly immunopositive for PLZF expression (black arrowhead), whereas only a small subset of stromal cells is PLZF negative (white arrowhead). The grey arrowhead indicates a glandular epithelial cell that is positive for PLZF expression. The scale bar in (D) applies to both panels (E,F).
Ijms 25 03451 g001
Figure 2. The PLZF transcription factor is not induced in the Plzf d/d uterus in response to acute progesterone exposure. (A) The schematic shows the P4 hormone treatment regimen for ovariectomized Plzf f/f control and Plzf d/d mutant mice that were used to induce uterine PLZF protein expression. Two weeks following ovariectomy, 8-week-old control and mutant mice received either P4 (1 mg) or hormone vehicle (sesame oil) through intrascapular s.c. injection. Mice were euthanized 12 h post-injection before uterine tissues were removed and processed for cellular and molecular analysis. (B) Shown are the PLZF immunoblot results for Plzf f/f mice treated with oil (lane 1), Plzf f/f mice treated with P4 (lane 2), Plzf d/d mice treated with oil (lane 3), and Plzf d/d mice treated with P4 (lane 4). Note: For each gel lane, uterine tissue protein was pooled from four mice per genotype and treatment group; β-actin served as a loading control. As expected, P4-induction of PLZF protein (~75 kDa) was detected only in the P4-treated Plzf f/f mouse group (lane 2). The absence of an immunoreactivity band for PLZF in the uterine protein preparation from the P4-treated Plzf d/d mouse group (lane 4) confirms that the engineered Plzf d/d mouse operates as designed. (C) Supporting the immunoblot data in panel (B), immunohistochemistry clearly detects PLZF protein expression in a subset of endometrial stromal (S) fibroblasts and luminal epithelial (LE) cells (black arrowhead) in the P4-treated Plzf f/f mouse (black arrowhead); stromal fibroblasts and epithelial cells that are PLZF negative are indicated by white arrowheads. (D) Confirming the immunoblot results in panel (B), endometrial PLZF protein is not detected by immunohistochemistry in the P4 treated Plzf d/d mouse (white arrowheads); the scale bar in (C) applies to panel (D). (E) Histograms display the results from the qRT-PCR analysis of expression from the following P4-responsive genes: Amphiregulin (Areg) [40]; Cytochrome P450 26A1 (Cyp26a1) [41]; ERBB receptor feedback inhibitor 1 (Errfi1) [42]; Heart and neural crest derivatives expressed 2 (Hand 2) [43]; Histidine decarboxylase (Hdc) [44]; Indian hedgehog (Ihh) [45]; Interleukin-13 receptor subunit alpha-2 (Il13ra2) [46]; promyelocytic leukemia zinc finger (Plzf) [33]; and Transcription factor 23 (Tcf 23) [47] in Plzf f/f and Plzf d/d uterine tissue following the P4 treatment described in panel (A). Note that for many of these P4-responsive genes, induction by P4 is either significantly attenuated or blocked in the Plzf d/d uterus (n = 4 mice per genotype and treatment). Not significant is denoted by n.s. whereas *, **, *** and **** indicate p values of <0.05, <0.01, <0.001 and <0.0001 respectively.
Figure 2. The PLZF transcription factor is not induced in the Plzf d/d uterus in response to acute progesterone exposure. (A) The schematic shows the P4 hormone treatment regimen for ovariectomized Plzf f/f control and Plzf d/d mutant mice that were used to induce uterine PLZF protein expression. Two weeks following ovariectomy, 8-week-old control and mutant mice received either P4 (1 mg) or hormone vehicle (sesame oil) through intrascapular s.c. injection. Mice were euthanized 12 h post-injection before uterine tissues were removed and processed for cellular and molecular analysis. (B) Shown are the PLZF immunoblot results for Plzf f/f mice treated with oil (lane 1), Plzf f/f mice treated with P4 (lane 2), Plzf d/d mice treated with oil (lane 3), and Plzf d/d mice treated with P4 (lane 4). Note: For each gel lane, uterine tissue protein was pooled from four mice per genotype and treatment group; β-actin served as a loading control. As expected, P4-induction of PLZF protein (~75 kDa) was detected only in the P4-treated Plzf f/f mouse group (lane 2). The absence of an immunoreactivity band for PLZF in the uterine protein preparation from the P4-treated Plzf d/d mouse group (lane 4) confirms that the engineered Plzf d/d mouse operates as designed. (C) Supporting the immunoblot data in panel (B), immunohistochemistry clearly detects PLZF protein expression in a subset of endometrial stromal (S) fibroblasts and luminal epithelial (LE) cells (black arrowhead) in the P4-treated Plzf f/f mouse (black arrowhead); stromal fibroblasts and epithelial cells that are PLZF negative are indicated by white arrowheads. (D) Confirming the immunoblot results in panel (B), endometrial PLZF protein is not detected by immunohistochemistry in the P4 treated Plzf d/d mouse (white arrowheads); the scale bar in (C) applies to panel (D). (E) Histograms display the results from the qRT-PCR analysis of expression from the following P4-responsive genes: Amphiregulin (Areg) [40]; Cytochrome P450 26A1 (Cyp26a1) [41]; ERBB receptor feedback inhibitor 1 (Errfi1) [42]; Heart and neural crest derivatives expressed 2 (Hand 2) [43]; Histidine decarboxylase (Hdc) [44]; Indian hedgehog (Ihh) [45]; Interleukin-13 receptor subunit alpha-2 (Il13ra2) [46]; promyelocytic leukemia zinc finger (Plzf) [33]; and Transcription factor 23 (Tcf 23) [47] in Plzf f/f and Plzf d/d uterine tissue following the P4 treatment described in panel (A). Note that for many of these P4-responsive genes, induction by P4 is either significantly attenuated or blocked in the Plzf d/d uterus (n = 4 mice per genotype and treatment). Not significant is denoted by n.s. whereas *, **, *** and **** indicate p values of <0.05, <0.01, <0.001 and <0.0001 respectively.
Ijms 25 03451 g002
Figure 3. Embryo implantation is impaired in the Plzf d/d mouse. (A) The left panel shows the gross morphology of the uterus obtained from the Plzf f/f control mouse at GD5. The Plzf f/f mouse received a tail vein injection of a Chicago Sky Blue dye solution minutes prior to euthanasia and tissue harvesting. Note the locations of implantation sites along each uterine horn as indicated by the intense Chicago Sky Blue staining (white arrowheads); the position of the stained ovaries is indicated by O. The right panel shows the gross morphology of the uterus derived from the similarly hormone treated Plzf d/d mutant at GD5. Note the clear absence of implantation sites along both Plzf d/d uterine horns, with only the ovaries scoring positive for Chicago Sky Blue staining. The histogram on the far right quantitatively displays the mean number of implantation sites per mouse in the Plzf f/f control group (n = 8); no implantation sites were observed in the Plzf d/d mutant group (n = 9). (B) The left panel shows a transverse uterine tissue section with an embryo from a Plzf f/f mouse at GD5; the tissue section is stained with H&E. Note the typical tight attachment of the embryo to the apical surface of the epithelial lining of the Plzf f/f endometrium (white arrowhead). The right panel shows a representative tissue section of a Plzf d/d uterus with an embryo at GD5, which is also stained with H&E. Note the absence of embryo implantation, but instead, the embryo floats in the open lumen (white arrowhead). (C) Quantitative real-time PCR analysis reveals that the expression of genes associated with murine uterine receptivity and P4-responsiveness (i.e., Bone morphogenetic protein 2 (Bmp 2 [49,50]) and Hand 2 [43]) is significantly attenuated in the Plzf d/d uterus when compared with the Plzf f/f uterus at GD5. Conversely, E2-responsive uterine biomarkers (i.e., Lactoferrin (Ltf) [51] and Early growth response 1 (Egr1) [33,52,53,54,55]) are significantly elevated in the Plzf d/d uterus when compared with the Plzf f/f uterus at GD5. Importantly, the levels of Estrogen receptor 1 (Esr1) and Pgr expression are not significantly altered between the Plzf f/f and Plzf d/d uteri at GD5. Not significant is denoted by ns whereas **, ***, and **** indicate p values of <0.01, <0.001, <0.0001 respectively.
Figure 3. Embryo implantation is impaired in the Plzf d/d mouse. (A) The left panel shows the gross morphology of the uterus obtained from the Plzf f/f control mouse at GD5. The Plzf f/f mouse received a tail vein injection of a Chicago Sky Blue dye solution minutes prior to euthanasia and tissue harvesting. Note the locations of implantation sites along each uterine horn as indicated by the intense Chicago Sky Blue staining (white arrowheads); the position of the stained ovaries is indicated by O. The right panel shows the gross morphology of the uterus derived from the similarly hormone treated Plzf d/d mutant at GD5. Note the clear absence of implantation sites along both Plzf d/d uterine horns, with only the ovaries scoring positive for Chicago Sky Blue staining. The histogram on the far right quantitatively displays the mean number of implantation sites per mouse in the Plzf f/f control group (n = 8); no implantation sites were observed in the Plzf d/d mutant group (n = 9). (B) The left panel shows a transverse uterine tissue section with an embryo from a Plzf f/f mouse at GD5; the tissue section is stained with H&E. Note the typical tight attachment of the embryo to the apical surface of the epithelial lining of the Plzf f/f endometrium (white arrowhead). The right panel shows a representative tissue section of a Plzf d/d uterus with an embryo at GD5, which is also stained with H&E. Note the absence of embryo implantation, but instead, the embryo floats in the open lumen (white arrowhead). (C) Quantitative real-time PCR analysis reveals that the expression of genes associated with murine uterine receptivity and P4-responsiveness (i.e., Bone morphogenetic protein 2 (Bmp 2 [49,50]) and Hand 2 [43]) is significantly attenuated in the Plzf d/d uterus when compared with the Plzf f/f uterus at GD5. Conversely, E2-responsive uterine biomarkers (i.e., Lactoferrin (Ltf) [51] and Early growth response 1 (Egr1) [33,52,53,54,55]) are significantly elevated in the Plzf d/d uterus when compared with the Plzf f/f uterus at GD5. Importantly, the levels of Estrogen receptor 1 (Esr1) and Pgr expression are not significantly altered between the Plzf f/f and Plzf d/d uteri at GD5. Not significant is denoted by ns whereas **, ***, and **** indicate p values of <0.01, <0.001, <0.0001 respectively.
Ijms 25 03451 g003
Figure 4. Uterine receptivity is impaired in the Plzf d/d mouse at GD4. (A) The left panels display the distribution of cells that are immunohistochemically positive for BrdU incorporation in the endometrium of the Plzf f/f mouse at GD4. The bottom panel is a higher magnification of a region shown in the top panel. Note that the majority of cells are BrdU negative in the luminal epithelium (LE), whereas a subset of stromal (S) cells is BrdU positive (black arrowhead), another subset of stromal cells is BrdU negative (white arrowhead). The right panels show the distribution of cells positive for BrdU incorporation in the endometrium of the Plzf d/d mouse at GD4. Again, the bottom panel shows a higher magnification of a region shown in the top panel. Note the retention of a significant number of BrdU-positive cells in the Plzf d/d luminal epithelium (black arrowhead). In addition, there is a marked decrease in the number of BrdU-positive stromal cells in the Plzf d/d endometrium (black arrowhead) at GD4; white arrowheads denote cells that are negative for BrdU. Note: scale bars in the left top and bottom panels also apply to the right top and bottom panels, respectively. (B) The histogram displays the quantitation of the relative mean number of BrdU-positive cells in the stromal and luminal epithelial compartments of the Plzf f/f and Plzf d/d uteri at GD4 (n = 4/genotype/cellular compartment); *** indicate a p value of <0.001.
Figure 4. Uterine receptivity is impaired in the Plzf d/d mouse at GD4. (A) The left panels display the distribution of cells that are immunohistochemically positive for BrdU incorporation in the endometrium of the Plzf f/f mouse at GD4. The bottom panel is a higher magnification of a region shown in the top panel. Note that the majority of cells are BrdU negative in the luminal epithelium (LE), whereas a subset of stromal (S) cells is BrdU positive (black arrowhead), another subset of stromal cells is BrdU negative (white arrowhead). The right panels show the distribution of cells positive for BrdU incorporation in the endometrium of the Plzf d/d mouse at GD4. Again, the bottom panel shows a higher magnification of a region shown in the top panel. Note the retention of a significant number of BrdU-positive cells in the Plzf d/d luminal epithelium (black arrowhead). In addition, there is a marked decrease in the number of BrdU-positive stromal cells in the Plzf d/d endometrium (black arrowhead) at GD4; white arrowheads denote cells that are negative for BrdU. Note: scale bars in the left top and bottom panels also apply to the right top and bottom panels, respectively. (B) The histogram displays the quantitation of the relative mean number of BrdU-positive cells in the stromal and luminal epithelial compartments of the Plzf f/f and Plzf d/d uteri at GD4 (n = 4/genotype/cellular compartment); *** indicate a p value of <0.001.
Ijms 25 03451 g004
Figure 5. Decidualization of the mouse uterus requires the PLZF transcription factor. (A) The schematic depicts the standard hormone treatment of ovariectomized Plzf f/f and Plzf d/d mice to prepare uterine tissue for an artificial decidual response following instillation of sesame oil (the deciduogenic stimulator) in one uterine horn. (B) The left panel shows the gross morphology of the Plzf f/f uterus following the hormone treatment regimen shown in panel (A). Note the appearance of the typical deciduomata in the stimulated (S) uterine horn, whereas the contralateral unstimulated (U) uterine horn does not display this tissue response. The right panel shows the similarly treated Plzf d/d uterus. Note the clear absence of a decidual response in the Plzf d/d uterine horn (S) that received the deciduogenic stimulus. (C) The histogram displays the means for the wet weight ratio of stimulated/unstimulated for Plzf f/f (n = 6) and Plzf d/d (n = 7) uteri in this study. (D) The left panel shows the transverse tissue section of the Plzf f/f stimulated (S) horn that is stained with H&E. The top right panel shows the corresponding unstimulated (U) uterine horn from the same Plzf f/f mouse. The bottom right panels show the stimulated (S) and unstimulated (U) uterine horns from the Plzf d/d mouse. Note that the size between the stimulated and unstimulated Plzf d/d horns is indistinguishable. The scale bar in the left panel applies to all panels in this panel series. (E) Quantitative real-time PCR analysis shows that typical gene expression levels associated with the uterine decidual response (i.e., Bmp2 [49,50], Hand2 [43], Prolactin family 3, subfamily c, member 1 (Prl3c1) [61] and Prolactin family 8, subfamily a, member 2 (Prl8a2) [62]) are significantly decreased in the Plzf d/d stimulated uterine horn when compared with the Plzf f/f stimulated horn; ** and *** indicate p values of <0.01 and <0.001 respectively (F) The tissue section of the Plzf f/f stimulated horn is stained for BrdU incorporation; the tissue section is located in the black square shown in panel (D) left panel. Note the presence of large decidual cells (white arrowhead), with only a few cells scoring positive for BrdU immunoreactivity (black arrowhead). (G) The tissue section stained for BrdU incorporation from the Plzf f/f stimulated uterine in a region indicated by the grey box in panel (D) left panel. Note the numerous cells positive for BrdU staining in the expanding cellular front of the deciduomata (black arrowhead). (H) The tissue section in the Plzf d/d stimulated uterine horn in a region indicated by the black box in panel (D) (bottom left panel); note that the majority of stromal cells are negative for BrdU incorporation, whereas the epithelial compartment still retains cells positive for BrdU (black arrowhead); LE, GE, and S denote luminal epithelium, glandular epithelium, and stroma, respectively. The scale bar in panel (F) also applies to panels (G,H).
Figure 5. Decidualization of the mouse uterus requires the PLZF transcription factor. (A) The schematic depicts the standard hormone treatment of ovariectomized Plzf f/f and Plzf d/d mice to prepare uterine tissue for an artificial decidual response following instillation of sesame oil (the deciduogenic stimulator) in one uterine horn. (B) The left panel shows the gross morphology of the Plzf f/f uterus following the hormone treatment regimen shown in panel (A). Note the appearance of the typical deciduomata in the stimulated (S) uterine horn, whereas the contralateral unstimulated (U) uterine horn does not display this tissue response. The right panel shows the similarly treated Plzf d/d uterus. Note the clear absence of a decidual response in the Plzf d/d uterine horn (S) that received the deciduogenic stimulus. (C) The histogram displays the means for the wet weight ratio of stimulated/unstimulated for Plzf f/f (n = 6) and Plzf d/d (n = 7) uteri in this study. (D) The left panel shows the transverse tissue section of the Plzf f/f stimulated (S) horn that is stained with H&E. The top right panel shows the corresponding unstimulated (U) uterine horn from the same Plzf f/f mouse. The bottom right panels show the stimulated (S) and unstimulated (U) uterine horns from the Plzf d/d mouse. Note that the size between the stimulated and unstimulated Plzf d/d horns is indistinguishable. The scale bar in the left panel applies to all panels in this panel series. (E) Quantitative real-time PCR analysis shows that typical gene expression levels associated with the uterine decidual response (i.e., Bmp2 [49,50], Hand2 [43], Prolactin family 3, subfamily c, member 1 (Prl3c1) [61] and Prolactin family 8, subfamily a, member 2 (Prl8a2) [62]) are significantly decreased in the Plzf d/d stimulated uterine horn when compared with the Plzf f/f stimulated horn; ** and *** indicate p values of <0.01 and <0.001 respectively (F) The tissue section of the Plzf f/f stimulated horn is stained for BrdU incorporation; the tissue section is located in the black square shown in panel (D) left panel. Note the presence of large decidual cells (white arrowhead), with only a few cells scoring positive for BrdU immunoreactivity (black arrowhead). (G) The tissue section stained for BrdU incorporation from the Plzf f/f stimulated uterine in a region indicated by the grey box in panel (D) left panel. Note the numerous cells positive for BrdU staining in the expanding cellular front of the deciduomata (black arrowhead). (H) The tissue section in the Plzf d/d stimulated uterine horn in a region indicated by the black box in panel (D) (bottom left panel); note that the majority of stromal cells are negative for BrdU incorporation, whereas the epithelial compartment still retains cells positive for BrdU (black arrowhead); LE, GE, and S denote luminal epithelium, glandular epithelium, and stroma, respectively. The scale bar in panel (F) also applies to panels (G,H).
Ijms 25 03451 g005
Table 1. Litter and pup number derived from Plzf f/f and Plzf d/d female mice after a six-month breeding period.
Table 1. Litter and pup number derived from Plzf f/f and Plzf d/d female mice after a six-month breeding period.
GenotypeNTotal Litter NumberTotal Pup NumberAverage Litter Number/DamAverage Pup Number/Dam
Plzf f/f13584584.435.0
Plzf d/d140000
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hai, L.; Maurya, V.K.; DeMayo, F.J.; Lydon, J.P. Establishment of Murine Pregnancy Requires the Promyelocytic Leukemia Zinc Finger Transcription Factor. Int. J. Mol. Sci. 2024, 25, 3451. https://doi.org/10.3390/ijms25063451

AMA Style

Hai L, Maurya VK, DeMayo FJ, Lydon JP. Establishment of Murine Pregnancy Requires the Promyelocytic Leukemia Zinc Finger Transcription Factor. International Journal of Molecular Sciences. 2024; 25(6):3451. https://doi.org/10.3390/ijms25063451

Chicago/Turabian Style

Hai, Lan, Vineet K. Maurya, Francesco J. DeMayo, and John P. Lydon. 2024. "Establishment of Murine Pregnancy Requires the Promyelocytic Leukemia Zinc Finger Transcription Factor" International Journal of Molecular Sciences 25, no. 6: 3451. https://doi.org/10.3390/ijms25063451

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop