Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Francesco J. DeMayo is active.

Publication


Featured researches published by Francesco J. DeMayo.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Forkhead box a2 (FOXA2) is essential for uterine function and fertility

Andrew M. Kelleher; Wang Peng; James K. Pru; Cindy A. Pru; Francesco J. DeMayo; Thomas E. Spencer

Significance These studies define biological roles for Forkhead box a2 (FOXA2) and glands of the uterus in female reproduction and fertility. FOXA2 is a critical regulator of uterine gland development in the neonate as well as of differentiated gland function in the adult uterus. Our findings provide clear in vivo evidence that FOXA2 regulates uterine expression of Leukemia inhibitory factor (Lif) for blastocyst implantation and that uterine glands play active roles in stromal cell decidualization and placental development. These findings support the idea that pregnancy loss and complications in women may have their origin in uterine gland dysfunction. Establishment of pregnancy is a critical event, and failure of embryo implantation and stromal decidualization in the uterus contribute to significant numbers of pregnancy losses in women. Glands of the uterus are essential for establishment of pregnancy in mice and likely in humans. Forkhead box a2 (FOXA2) is a transcription factor expressed specifically in the glands of the uterus and is a critical regulator of postnatal uterine gland differentiation in mice. In this study, we conditionally deleted FOXA2 in the adult mouse uterus using the lactotransferrin Cre (Ltf-Cre) model and in the neonatal mouse uterus using the progesterone receptor Cre (Pgr-Cre) model. The uteri of adult FOXA2-deleted mice were morphologically normal and contained glands, whereas the uteri of neonatal FOXA2-deleted mice were completely aglandular. Notably, adult FOXA2-deleted mice are completely infertile because of defects in blastocyst implantation and stromal cell decidualization. Leukemia inhibitory factor (LIF), a critical implantation factor of uterine gland origin, was not expressed during early pregnancy in adult FOXA2-deleted mice. Intriguingly, i.p. injections of LIF initiated blastocyst implantation in the uteri of both gland-containing and glandless adult FOXA2-deleted mice. Although pregnancy was rescued by LIF and was maintained to term in uterine gland-containing adult FOXA2-deleted mice, pregnancy failed by day 10 in neonatal FOXA2-deleted mice lacking uterine glands. These studies reveal a previously unrecognized role for FOXA2 in regulation of adult uterine function and fertility and provide original evidence that uterine glands and, by inference, their secretions play important roles in blastocyst implantation and stromal cell decidualization.


Biology of Reproduction | 2017

Decreased epithelial progesterone receptor A at the window of receptivity is required for preparation of the endometrium for embryo attachment

Margeaux Wetendorf; San-Pin Wu; Xiaoqiu Wang; Chad J. Creighton; Tianyuan Wang; Rainer B. Lanz; Leen J. Blok; Sophia Y. Tsai; Ming-Jer Tsai; John P. Lydon; Francesco J. DeMayo

Abstract The precise timing of progesterone signaling through its cognate receptor, the progesterone receptor (PGR), is critical for the establishment and maintenance of pregnancy. Loss of PGR expression in the murine uterine epithelium during the preimplantation period is a marker for uterine receptivity and embryo attachment. We hypothesized that the decrease in progesterone receptor A (PGRA) expression is necessary for successful embryo implantation. To test this hypothesis, a mouse model constitutively expressing PGRA (mPgrA LsL/+) was generated. Expression of PGRA in all uterine compartments (Pgrcre ) or uterine epithelium (Wnt7acre ) resulted in infertility with defects in embryo attachment and stromal decidualization. Expression of critical PGRA target genes, indian hedgehog, and amphiregulin (Areg), wasmaintained through the window of receptivity while the estrogen receptor target gene, the leukemia inhibitory factor (Lif), a key regulator of embryo receptivity, was decreased. Transcriptomic and cistromic analyses of the mouse uterus at day 4.5 of pregnancy identified an altered group of genes regulating molecular transport in the control of fluid and ion levels within the uterine interstitial space. Additionally, LIF and its cognate receptor, the leukemia inhibitory factor receptor (LIFR), exhibited PGR-binding events in regions upstream of the transcriptional start sites, suggesting PGRA is inhibiting transcription at these loci. Therefore, downregulation of the PGRA isoform at the window of receptivity is necessary for the attenuation of hedgehog signaling, transcriptional activation of LIF signaling, and modulation of solutes and fluid, producing a receptive environment for the attaching embryo. Summary Sentence Expression of PGRA at the window of receptivity transcriptionally represses LIF signaling and aberrantly regulates hedgehog and solute signaling rendering the uterus unreceptive to the implanting embryo.


Current Topics in Developmental Biology | 2017

Chapter Six – Progesterone Receptor Signaling in Uterine Myometrial Physiology and Preterm Birth

San-Pin Wu; Francesco J. DeMayo

Myometrium holds the structural integrity for the uterus and generates force for parturition with its primary component, the smooth muscle cells. The progesterone receptor mediates progesterone-dependent signaling and connects to a network of pathways for regulation of contractility and inflammatory responses in myometrium. Dysfunctional progesterone signaling has been linked to pregnancy complications including preterm birth. In the present review, we summarize recent findings on modifiers and effectors of the progesterone receptor signaling. Discussions include novel conceptual discoveries and new development in legacy pathways such as the signal transducers NF-κB, ZEB, microRNA, and the unfolded protein response pathways. We also discuss the impact of progesterone receptor isoform composition and ligand accessibility in modification of the progesterone receptor genomic actions.


Molecular Human Reproduction | 2017

Growth regulation by estrogen in breast cancer 1 (GREB1) is a novel progesterone-responsive gene required for human endometrial stromal decidualization

Alison J. Camden; Maria M. Szwarc; Sangappa B. Chadchan; Francesco J. DeMayo; Bert W. O'Malley; John P. Lydon; Ramakrishna Kommagani

STUDY QUESTION Is Growth Regulation by Estrogen in Breast Cancer 1 (GREB1) required for progesterone-driven endometrial stromal cell decidualization? SUMMARY ANSWER GREB1 is a novel progesterone-responsive gene required for progesterone-driven human endometrial stromal cell (HESC) decidualization. WHAT IS KNOWN ALREADY Successful establishment of pregnancy requires HESCs to transform from fibroblastic to epithelioid cells in a process called decidualization. This process depends on the hormone progesterone, but the molecular mechanisms by which it occurs have not been determined. STUDY DESIGN, SIZE, DURATION Primary and transformed HESCs in which GREB1 expression was knocked down were decidualized in culture for up to 6 days. Wild-type and progesterone receptor (PR) knockout mice were treated with progesterone, and their uteri were assessed for levels of GREB1 expression. PARTICIPANTS/MATERIALS, SETTING, METHODS Analysis of previous data included data mining of expression profile data sets and in silico transcription factor-binding analysis. Endometrial biopsies obtained from healthy women of reproductive age during the proliferative phase (Days 8-12) of their menstrual cycle were used for isolating HESCs. Experiments were carried out with early passage (no more than four passages) HESCs isolated from at least three subjects. Transcript levels of decidualization markers prolactin (PRL) and insulin-like growth factor-binding protein-1 (IGFBP-1) were detected by quantitative RT-PCR as readouts for HESC decidualization. Cells were also imaged by phase-contrast microscopy. To assess the requirement for GREB1, PR and SRC-2, cells were transfected with specifically targeted small interfering RNAs. Results are shown as mean and SE from three replicates of one representative patient-derived primary endometrial cell line. Experiments were also conducted with transformed HESCs. MAIN RESULTS AND THE ROLE OF CHANCE Progesterone treatment of mice and transformed HESCs led to an ~5-fold (5.6 ± 0.81, P < 0.05, and 5.2 ± 0.26, P < 0.01, respectively) increase in GREB1 transcript levels. This increase was significantly reduced in the uteri of PR knock-out mice (P < 0.01), in HESCs treated with the PR antagonist RU486 (P < 0.01), or in HESCs in which PR expression was knocked down (P < 0.05). When GREB1 expression was knocked down, progesterone-driven decidualization markers in both immortalized and primary HESCs was significantly reduced (P < 0.05 and P < 0.01). Finally, GREB1 knock down signficantly reduced expression of the PR target genes WNT4 and FOXOA1 (P < 0.05 and P < 0.01, respectively). LARGE SCALE DATA This study used the Nuclear Receptor Signaling Atlas. LIMITATIONS, REASONS FOR CAUTION Although in vitro cell culture studies indicate that GREB1 is required for endoemtrial decidualization, the in vivo role of GREB1 in endometrial function and dysfunction should be assessed by using knock-out mouse models. WIDER IMPLICATIONS OF THE FINDINGS Identification and functional analysis of GREB1 as a key molecular mediator of decidualization may lead to improved diagnosis and clinical management of women with peri-implantation loss due to inadequate endometrial decidualization. STUDY FUNDING AND COMPETING INTEREST(S) This research was funded in part by: a National Institutes of Health (NIH)/ National Institute of Child Health and Human Development (NICHD) grant (R00 HD080742) and Washington University School of Medicine start-up funds to R.K., an NIH/NICHD grant (RO1 HD-07857) to B.W.O.M., and a NIH/NICHD grant (R01 HD-042311) to J.P.L. The authors declare no conflicts of interests.


Endocrinology | 2017

Deletion of RhoA in Progesterone Receptor–Expressing Cells Leads to Luteal Insufficiency and Infertility in Female Mice

Ahmed E. El Zowalaty; Rong Li; Yi Zheng; John P. Lydon; Francesco J. DeMayo; Xiaoqin Ye

Ras homolog gene family, member A (RhoA) is widely expressed throughout the female reproductive system. To assess its role in progesterone receptor-expressing cells, we generated RhoA conditional knockout mice RhoAd/d (RhoAf/f-Pgr-Cre+/−). RhoAd/d female mice had comparable mating activity, serum luteinizing hormone, prolactin, and estradiol levels and ovulation with control but were infertile with progesterone insufficiency, indicating impaired steroidogenesis in RhoAd/d corpus luteum (CL). RhoA was highly expressed in wild-type luteal cells and conditionally deleted in RhoAd/d CL. Gestation day 3.5 (D3.5) RhoAd/d ovaries had reduced numbers of CL, less defined corpus luteal cord formation, and disorganized CL collagen IV staining. RhoAd/d CL had lipid droplet and free cholesterol accumulation, indicating the availability of cholesterol for steroidogenesis, but disorganized β-actin and vimentin staining, indicating disrupted cytoskeleton integrity. Cytoskeleton is important for cytoplasmic cholesterol movement to mitochondria and for regulating mitochondria. Dramatically reduced expression of mitochondrial markers heat shock protein 60 (HSP60), voltage-dependent anion channel, and StAR was detected in RhoAd/d CL. StAR carries out the rate-limiting step of steroidogenesis. StAR messenger RNA expression was reduced in RU486-treated D3.5 wild-type CL and tended to be induced in progesterone-treated D3.5 RhoAd/d CL, with parallel changes of HSP60 expression. These data demonstrated the in vivo function of RhoA in CL luteal cell cytoskeleton integrity, cholesterol transport, StAR expression, and progesterone synthesis, and a positive feedback on StAR expression in CL by progesterone signaling. These findings provide insights into mechanisms of progesterone insufficiency.


Biology of Reproduction | 2018

Uterine function in the mouse requires speckle-type poz protein

Lan Hai; Maria M. Szwarc; Bin He; David M. Lonard; Ramakrishna Kommagani; Francesco J. DeMayo; John P. Lydon

Abstract Speckle-type poz protein (SPOP) is an E3-ubiquitin ligase adaptor for turnover of a diverse number of proteins involved in key cellular processes such as chromatin remodeling, transcriptional regulation, and cell signaling. Genomic analysis revealed that SPOP somatic mutations are found in a subset of endometrial cancers, suggesting that these mutations act as oncogenic drivers of this gynecologic malignancy. These studies also raise the question as to the role of wild-type SPOP in normal uterine function. To address this question, we generated a mouse model (Spopd/d) in which SPOP is ablated in uterine cells that express the PGR. Fertility studies demonstrated that SPOP is required for embryo implantation and for endometrial decidualization. Molecular analysis revealed that expression levels of the PGR at the protein and transcript level are significantly reduced in the Spopd/d uterus. While this result was unexpected, this finding explains in part the dysfunctional phenotype of the Spopd/d uterus. Moderate increased levels of the ESR1, GATA2, and SRC2 were detected in the Spopd/d uterus, suggesting that SPOP is required to maintain the proteome for normal uterine function. With age, the Spopd/d endometrium exhibits large glandular cysts with foci of epithelial proliferation, further supporting a role for SPOP in maintaining a healthy uterus. Collectively, studies on the Spopd/d mouse support an important role for SPOP in normal uterine function and suggest that this mouse model may prove useful to study the role of SPOP-loss-of-function mutations in the etiopathogenesis of endometrial cancer. Summary Sentence SPOP is required for embryo implantation, endometrial decidualization, and uterine health in the mouse


Biology of Reproduction | 2018

Human endometrial stromal cell decidualization requires transcriptional reprogramming by PLZF

Maria M. Szwarc; Lan Hai; William E. Gibbons; M. Peavey; Lisa D. White; Qianxing Mo; David M. Lonard; Ramakrishna Kommagani; Rainer B. Lanz; Francesco J. DeMayo; John P. Lydon

Abstract Infertility and early embryo miscarriage is linked to inadequate endometrial decidualization. Although transcriptional reprogramming is known to drive decidualization in response to progesterone, the key signaling effectors that directlymediate this hormone response are not fully known. This knowledge gap is clinically significant because identifying the early signals that directly mediate progesterone-driven decidualization will address some of the current limitations in diagnosing and therapeutically treating patients at most risk for early pregnancy loss.We recently revealed that the promyelocytic leukemia zinc finger (PLZF) is a direct target of the progesterone receptor and is essential for decidualization of human endometrial stromal cells (hESCs). The purpose of this current work was to identify the genome-wide transcriptional program that is controlled by PLZF during hESC decidualization using an established in vitro hESC culture model, siRNA-mediated knockdownmethods, and RNA-sequencing technology followed by bioinformatic analysis and validation. We discovered that PLZF is critical in the regulation of genes that are involved in cellular processes that are essential for the archetypal morphological and functional changes that occur when hESCs transform into epithelioid decidual cells such as proliferation and cell motility. We predict that the transcriptome datasets identified in this study will not only contribute to a broader understanding of PLZF-dependent endometrial decidualization at the molecular level but may advance the development of more effective molecular diagnostics and therapeutics for the clinical management of female infertility and subfertility that is based on a dysfunctional endometrium. Summary Sentence PLZF drives progesterone-dependent transcriptional reprogramming of the human endometrial stromal cells to enable decidualization.


BMC Biology | 2018

Comparative analysis of single-stranded DNA donors to generate conditional null mouse alleles

Denise G. Lanza; Angelina Gaspero; Isabel Lorenzo; Lan Liao; Ping Zheng; Ying Wang; Yu Deng; Chonghui Cheng; Chuansheng Zhang; John R. Seavitt; Francesco J. DeMayo; Jianming Xu; Mary E. Dickinson; Arthur L. Beaudet; Jason D. Heaney

BackgroundThe International Mouse Phenotyping Consortium is generating null allele mice for every protein-coding gene in the genome and characterizing these mice to identify gene–phenotype associations. While CRISPR/Cas9-mediated null allele production in mice is highly efficient, generation of conditional alleles has proven to be more difficult. To test the feasibility of using CRISPR/Cas9 gene editing to generate conditional knockout mice for this large-scale resource, we employed Cas9-initiated homology-driven repair (HDR) with short and long single stranded oligodeoxynucleotides (ssODNs and lssDNAs).ResultsUsing pairs of single guide RNAs and short ssODNs to introduce loxP sites around a critical exon or exons, we obtained putative conditional allele founder mice, harboring both loxP sites, for 23 out of 30 targeted genes. LoxP sites integrated in cis in at least one mouse for 18 of 23 genes. However, loxP sites were mutagenized in 4 of the 18 in cis lines. HDR efficiency correlated with Cas9 cutting efficiency but was minimally influenced by ssODN homology arm symmetry. By contrast, using pairs of guides and single lssDNAs to introduce loxP-flanked exons, conditional allele founders were generated for all four genes targeted, although one founder was found to harbor undesired mutations within the lssDNA sequence interval. Importantly, when employing either ssODNs or lssDNAs, random integration events were detected.ConclusionsOur studies demonstrate that Cas9-mediated HDR with pairs of ssODNs can generate conditional null alleles at many loci, but reveal inefficiencies when applied at scale. In contrast, lssDNAs are amenable to high-throughput production of conditional alleles when they can be employed. Regardless of the single-stranded donor utilized, it is essential to screen for sequence errors at sites of HDR and random insertion of donor sequences into the genome.


Placenta | 2017

Hormone dependent uterine epithelial-stromal communication for pregnancy support

Xiaoqiu Wang; San-Pin Wu; Francesco J. DeMayo

Human fertility is a relatively inefficient process. Despite the presence of visibly healthy embryos, 30% of pregnancies generated by assisted reproductive technology (ART) fail before the second trimester. The uterine microenvironment plays a critical role in establishing and maintaining a successful pregnancy that requires coordinated communication between the epithelial and stromal cells of the endometrium. The epithelial cells must cease proliferation and become permissive for the conceptus (embryo and associated extraembryonic membranes), while the stromal cells undergoes mesenchymal-to-epithelioid transformation to form the decidua in support of subsequent embryo development. The ovarian steroids Estrogen (E2) and Progesterone (P4) are the major hormones governing these processes. These hormones act via their nuclear receptors, the estrogen receptor, ESR1, and progesterone receptor, PGR, to direct the transcription of genes that orchestrate epithelial and stromal cell communication. This review will discuss the molecular mechanisms utilized by steroid hormones that regulate uterine receptivity, as well, establish and maintain pregnancy.


Lung Cancer | 2017

Mig-6 deficiency cooperates with oncogenic Kras to promote mouse lung tumorigenesis

Jian Liu; Sung Nam Cho; San Pin Wu; Nili Jin; Seyed Javad Moghaddam; Jennifer L. Gilbert; Ignacio I. Wistuba; Francesco J. DeMayo

OBJECTIVES Lung cancer is the leading cause of cancer related deaths worldwide and mutation activating KRAS is one of the most frequent mutations found in lung adenocarcinoma. Identifying regulators of KRAS may aid in the development of therapies to treat this disease. The mitogen-induced gene 6, MIG-6, is a small adaptor protein modulating signaling in cells to regulate the growth and differentiation in multiple tissues. Here, we investigated the role of Mig-6 in regulating adenocarcinoma progression in the lungs of genetically engineered mice with activation of Kras. MATERIALS AND METHODS Using the CCSPCre mouse to specifically activate expression of the oncogenic KrasG12D in Club cells, we investigated the expression of Mig-6 in CCSPCreKrasG12D-induced lung tumors. To determine the role of Mig-6 in KrasG12D-induced lung tumorigenesis, Mig-6 was conditionally ablated in the Club cells by breeding Mig6f/f mice to CCSPCreKrasG12D mice, yielding CCSPCreMig-6d/dKrasG12D mice (Mig-6d/dKrasG12D). RESULTS We found that Mig-6 expression is decreased in CCSPCreKrasG12D-induced lung tumors. Ablation of Mig-6 in the KrasG12D background led to enhanced tumorigenesis and reduced life expectancy. During tumor progression, there was increased airway hyperplasia, a heightened inflammatory response, reduced apoptosis in KrasG12D mouse lungs, and an increase of total and phosphorylated ERBB4 protein levels. Mechanistically, Mig-6 deficiency attenuates the cell apoptosis of lung tumor expressing KRASG12D partially through activating the ErbB4 pathway. CONCLUSIONS In summary, Mig-6 deficiency promotes the development of KrasG12D-induced lung adenoma through reducing the cell apoptosis in KrasG12D mouse lungs partially by activating the ErbB4 pathway.

Collaboration


Dive into the Francesco J. DeMayo's collaboration.

Top Co-Authors

Avatar

John P. Lydon

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

San-Pin Wu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Maria M. Szwarc

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Rainer B. Lanz

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ramakrishna Kommagani

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ming-Jer Tsai

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lan Hai

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jian Liu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

M. Peavey

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge