Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 27(5): 1489-1504, 2020 05.
Article in English | MEDLINE | ID: mdl-31595043

ABSTRACT

A reciprocal communication between the implantation-competent blastocyst and the receptive uterus is essential to successful implantation and pregnancy success. Progesterone (P4) signaling via nuclear progesterone receptor (PR) is absolutely critical for pregnancy initiation and its success in most eutherian mammals. Here we show that a nuclear protein high-mobility group box-1 (HMGB1) plays a critical role in implantation in mice by preserving P4-PR signaling. Conditional deletion of uterine Hmgb1 by a Pgr-Cre driver shows implantation defects accompanied by decreased stromal cell Hoxa10 expression and cell proliferation, two known signatures of inefficient responsiveness of stromal cells to PR signaling in implantation. These mice evoke inflammatory conditions with sustained macrophage accumulation in the stromal compartment on day 4 of pregnancy with elevated levels of macrophage attractants Csf1 and Ccl2. The results are consistent with the failure of exogenous P4 administration to rescue implantation deficiency in the mutant females. These early defects are propagated throughout the course of pregnancy and ultimately result in substantial subfertility. Collectively, the present study provides evidence that nuclear HMGB1 contributes to successful blastocyst implantation by sustaining P4-PR signaling and restricting macrophage accumulation to attenuate harmful inflammatory responses.


Subject(s)
HMGB1 Protein/deficiency , Uterus/metabolism , Uterus/pathology , Animals , Cytokines/metabolism , Decidua/pathology , Embryo Implantation , Female , Gene Deletion , HMGB1 Protein/metabolism , Infertility, Female/pathology , Macrophages/metabolism , Male , Mice , Pregnancy , Pregnancy Outcome , Receptors, Progesterone/metabolism , Stromal Cells/metabolism
2.
Nat Commun ; 10(1): 5425, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31780662

ABSTRACT

Scribble (Scrib) is a scaffold protein with multifunctional roles in PCP, tight junction and Hippo signaling. This study shows that Scrib is expressed in stromal cells around the implantation chamber following implantation. Stromal cells transform into epithelial-like cells to form the avascular primary decidual zone (PDZ) around the implantation chamber (crypt). The PDZ creates a permeability barrier around the crypt restricting immune cells and harmful agents from maternal circulation to protect embryonic health. The mechanism underlying PDZ formation is not yet known. We found that uterine deletion of Scrib by a Pgr-Cre driver leads to defective PDZ formation and implantation chamber (crypt) formation, compromising pregnancy success. Interestingly, epithelial-specific Scrib deletion by a lactoferrin-Cre (Ltf-Cre) driver does not adversely affect PDZ formation and pregnancy success. These findings provide evidence for a previously unknown function of stromal Scrib in PDZ formation, potentially involving ZO-1 and Hippo signaling.


Subject(s)
Decidua/metabolism , Embryo Implantation/genetics , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Stromal Cells/metabolism , Animals , Decidua/cytology , Epithelial Cells/cytology , Female , Gene Deletion , Hippo Signaling Pathway , Mice , Pregnancy , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Stromal Cells/cytology , Uterus/cytology , Uterus/metabolism , Zonula Occludens-1 Protein/metabolism
3.
Cell Rep ; 27(6): 1755-1768.e4, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31067461

ABSTRACT

Preterm birth (PTB) is a syndrome with many origins. Among them, infection or inflammation are major risk factors for PTB; however, local defense mechanisms to mount anti-inflammatory responses against inflammation-induced PTB are poorly understood. Here, we show that endothelial TLR4 in the decidual bed is critical for sensing inflammation during pregnancy because mice with endothelial Tlr4 deletion are resistant to lipopolysaccharide (LPS)-induced PTB. Under inflammatory conditions, IL-6 is readily expressed in decidual endothelial cells with signal transducer and activator of transcription 3 (Stat3) phosphorylation in perivascular stromal cells, which then regulates expression of anti-inflammatory IL-10. Our observation that administration of an IL-10 neutralizing antibody predisposing mice to PTB shows IL-10's anti-inflammatory role to prevent PTB. We show that the integration of endothelial and perivascular stromal signaling can determine pregnancy outcomes. These findings highlight a role for endothelial TLR4 in inflammation-induced PTB and may offer a potential therapeutic target to prevent PTB.


Subject(s)
Decidua/pathology , Endothelial Cells/metabolism , Molecular Targeted Therapy , Premature Birth/pathology , Premature Birth/prevention & control , Animals , Cell Communication/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Gene Deletion , Inflammation/pathology , Interleukin-10/metabolism , Interleukin-6/pharmacology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides , Mice , NF-kappa B/metabolism , Ovary/metabolism , Pregnancy , STAT3 Transcription Factor/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Toll-Like Receptor 4/metabolism
4.
Proc Natl Acad Sci U S A ; 113(50): E8079-E8088, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911818

ABSTRACT

Blastocyst implantation is a complex process requiring coordination of a dynamic sequence of embryo-uterine interactions. Blood vessels enter the uterus from the mesometrium, demarcating the uterus into mesometrial (M) and antimesometrial (AM) domains. Implantation occurs along the uterine longitudinal axis within specialized implantation chambers (crypts) that originate within the evaginations directed from the primary lumen toward the AM domain. The morphological orientation of crypts in rodent uteri was recognized more than a century ago, but the mechanism remained unknown. Here we provide evidence that planar cell polarity (PCP) signaling orchestrates directed epithelial evaginations to form crypts for implantation in mice. Uterine deletion of Vang-like protein 2, but not Vang-like protein 1, conferred aberrant PCP signaling, misdirected epithelial evaginations, defective crypt formation, and blastocyst attachment, leading to severely compromised pregnancy outcomes. The study reveals a previously unrecognized role for PCP in executing spatial cues for crypt formation and implantation. Because PCP is an evolutionarily conserved phenomenon, our study is likely to inspire implantation studies of this signaling pathway in humans and other species.


Subject(s)
Cell Polarity/physiology , Embryo Implantation/physiology , Uterus/physiology , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Communication/physiology , Dishevelled Proteins/physiology , Epithelium/anatomy & histology , Epithelium/physiology , Female , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Pregnancy , Pregnancy Outcome , Receptor Tyrosine Kinase-like Orphan Receptors/deficiency , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/physiology , Signal Transduction/physiology , Uterus/anatomy & histology , Wnt-5a Protein/deficiency , Wnt-5a Protein/genetics , Wnt-5a Protein/physiology
5.
J Clin Invest ; 126(8): 2941-54, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27454290

ABSTRACT

Inflammation and oxidative stress are known risk factors for preterm birth (PTB); however, the mechanisms and pathways that influence this condition are not fully described. Previously, we showed that mTORC1 signaling is increased in mice harboring a uterine-specific deletion of transformation-related protein 53 (p53d/d mice), which exhibit premature decidual senescence that triggers spontaneous and inflammation-induced PTB. Treatment with the mTORC1 inhibitor rapamycin reduced the incidence of PTB in the p53d/d mice. Decidual senescence with heightened mTORC1 signaling is also a signature of human PTB. Here, we have identified an underlying mechanism for PTB and a potential therapeutic strategy for treating the condition. Treatment of pregnant p53d/d mice with either the antidiabetic drug metformin or the antioxidant resveratrol activated AMPK signaling and inhibited mTORC1 signaling in decidual cells. Both metformin and resveratrol protected against spontaneous and inflammation-induced PTB in p53d/d females. Using multiple approaches, we determined that p53 interacts with sestrins to coordinate an inverse relationship between AMPK and mTORC1 signaling that determines parturition timing. This signature was also observed in human decidual cells. Together, these results reveal that p53-dependent coordination of AMPK and mTORC1 signaling controls parturition timing and suggest that metformin and resveratrol have therapeutic potential to prevent PTB.


Subject(s)
AMP-Activated Protein Kinases/physiology , Decidua/physiology , Multiprotein Complexes/physiology , Nuclear Proteins/physiology , Parturition/physiology , TOR Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/physiology , Animals , Antioxidants/metabolism , Binding Sites , Cellular Senescence , Female , Humans , Inflammation , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Oxidative Stress , Peroxidases , Pregnancy , Premature Birth , Signal Transduction , Up-Regulation , Uterus/physiology
6.
JCI Insight ; 1(3)2016 Mar 17.
Article in English | MEDLINE | ID: mdl-27159542

ABSTRACT

Preeclampsia (PE) is a disorder of pregnancy that manifests as late gestational maternal hypertension and proteinuria and can be life-threatening to both the mother and baby. It is believed that abnormal placentation is responsible for the cascade of events leading to the maternal syndrome. Embryo implantation is critical to establishing a healthy pregnancy. Defective implantation can cause adverse "ripple effects," leading to abnormal decidualization and placentation, retarded fetal development, and poor pregnancy outcomes, such as PE and fetal growth restriction. The precise mechanism(s) of implantation defects that lead to PE remain elusive. BPH/5 mice, which spontaneously develop the cardinal features of PE, show peri-implantation defects including upregulation of Cox2 and IL-15 at the maternal-fetal interface. This was associated with decreased decidual natural killer (dNK) cells, which have important roles in establishing placental perfusion. Interestingly, a single administration of a Cox2 inhibitor (celecoxib) during decidualization restrained Cox2 and IL-15 expression, restored dNK cell numbers, improved fetal growth, and attenuated late gestational hypertension in BPH/5 female mice. This study provides evidence that decidual overexpression of Cox2 and IL-15 may trigger the adverse pregnancy outcomes reflected in the preeclamptic syndrome, underscoring the idea that Cox2 inhibitor treatment is an effective strategy for the prevention of PE-associated fetal and maternal morbidity and mortality.

7.
J Biol Chem ; 290(24): 15337-49, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25931120

ABSTRACT

Embryonic diapause is a reproductive strategy widespread in the animal kingdom. This phenomenon is defined by a temporary arrest in blastocyst growth and metabolic activity within a quiescent uterus without implantation until the environmental and maternal milieu become favorable for pregnancy to progress. We found that uterine Msx expression persists during diapause across species; their inactivation in the mouse uterus results in termination of diapause with the development of implantation-like responses ("pseudoimplantation") that ultimately succumbed to resorption. To understand the cause of this failure, we compared proteome profiles between floxed and Msx-deleted uteri. In deleted uteri, several functional networks, including transcription/translation, ubiquitin-proteasome, inflammation, and endoplasmic reticulum stress, were dysregulated. Computational modeling predicted intersection of these pathways on an enhanced inflammatory signature. Further studies showed that this signature was reflected in increased phosphorylated IκB levels and nuclear NFκB in deleted uteri. This was associated with enhanced proteasome activity and endoplasmic reticulum stress. Interestingly, treatment with anti-inflammatory glucocorticoid (dexamethasone) reduced the inflammatory signature with improvement of the diapause phenotype. These findings highlight an unexpected role of uterine Msx in limiting aberrant inflammatory responses to maintain embryonic diapause.


Subject(s)
Embryonic Development/genetics , Genes, Homeobox , Homeodomain Proteins/physiology , Inflammation/genetics , MSX1 Transcription Factor/physiology , Muscle, Smooth/pathology , Uterus/pathology , Animals , Female , Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Mice , Mice, Knockout , Pregnancy , Uterus/metabolism
8.
Physiol Rep ; 3(4)2015 Apr.
Article in English | MEDLINE | ID: mdl-25896980

ABSTRACT

We investigated the role of muscle activity in maintaining normal glucose homeostasis via transection of the sciatic nerve, an extreme model of disuse atrophy. Mice were killed 3, 10, 28, or 56 days after transection or sham surgery. There was no difference in muscle weight between sham and transected limbs at 3 days post surgery, but it was significantly lower following transection at the other three time points. Transected muscle weight stabilized by 28 days post surgery with no further loss. Myocellular cross-sectional area was significantly smaller at 10, 28, and 56 days post transection surgery. Additionally, muscle fibrosis area was significantly greater at 56 days post transection. In transected muscle there was reduced expression of genes encoding transcriptional regulators of metabolism (PPARα, PGC-1α, PGC-1ß, PPARδ), a glycolytic enzyme (PFK), a fatty acid transporter (M-CPT 1), and an enzyme of mitochondrial oxidation (CS) with transection. In denervated muscle, glucose uptake was significantly lower at 3 days but was greater at 56 days under basal and insulin-stimulated conditions. Although GLUT 4 mRNA was significantly lower at all time points in transected muscle, Western blot analysis showed greater expression of GLUT4 at 28 and 56 days post surgery. GLUT1 mRNA was unchanged; however, GLUT1 protein expression was also greater in transected muscles. Surgery led to significantly higher protein expression for Akt2 as well as higher phosphorylation of Akt. While denervation may initially lead to reduced glucose sensitivity, compensatory responses of insulin signaling appeared to restore and improve glucose uptake in long-term-transected muscle.

9.
Cell Rep ; 8(2): 382-92, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25043182

ABSTRACT

Embryo homing and implantation occur within a crypt (implantation chamber) at the antimesometrial (AM) pole along the uterus. The mechanism by which this is achieved is not known. Here, we show that villi-like epithelial projections from the main uterine lumen toward the AM pole at regularly spaced intervals that form crypts for embryo implantation were disrupted in mice with uterine loss or gain of function of Wnt5a, or loss of function of both Ror1 and Ror2. This disruption of Wnt5a-ROR signaling resulted in disorderly epithelial projections, crypt formation, embryo spacing, and impaired implantation. These early disturbances under abnormal Wnt5a-ROR signaling were reflected in adverse late pregnancy events, including defective decidualization and placentation, ultimately leading to compromised pregnancy outcomes. This study presents deeper insight regarding the formation of organized epithelial projections for crypt formation and embryo implantation for pregnancy success.


Subject(s)
Decidua/metabolism , Embryo Implantation , Epithelial Cells/cytology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Decidua/cytology , Decidua/physiology , Epithelial Cells/metabolism , Female , Mice , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Wnt Proteins/genetics , Wnt-5a Protein
10.
Mol Endocrinol ; 28(7): 1108-17, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24850415

ABSTRACT

Although cervical pregnancy and placenta previa, in which the embryo and placenta embed in or adjacent to the cervix, are life-threatening complications that result in massive bleeding and poor pregnancy outcomes in women, the incidence of these aberrant conditions is uncommon. We hypothesized that a local molecular mechanism is normally in place to prevent embryo implantation in the cervix. The ovarian hormones progesterone (P(4)) and estrogen differentially direct differentiation and proliferation of endometrial cells, which confers the receptive state for implantation: P(4) dominance causes differentiation of the luminal epithelium but increases stromal cell proliferation in preparation of the uterus for implantation. In search for the cause of cervical nonresponsiveness to implantation, we found that the statuses of cell proliferation and differentiation between the uterus and cervix during early pregnancy are remarkably disparate under identical endocrine milieu in both mice and humans. We also found that cervical levels of progesterone receptor (PR) protein are low compared with uterine levels during this period, and the low PR protein levels are attributed to elevated levels of microRNA(miR)-200a in the cervix. These changes were associated with up-regulation of the P(4)-metabolizing enzyme 20α-hydroxysteroid dehydrogenase (200α-HSD) and down-regulation of its transcriptional repressor signal transducer and activator of transcription 5 in the cervix. The results provide evidence that elevated levels of miR-200a lead to down-regulation of P(4)-PR signaling and up-regulation of (200α-HSD) in the cervix, rendering it nonresponsive to implantation. These findings may point toward not only the physiological but also the pathological basis of the cervical milieu in embryo implantation.


Subject(s)
20-alpha-Hydroxysteroid Dehydrogenase/biosynthesis , Cervix Uteri/metabolism , Embryo Implantation, Delayed/genetics , MicroRNAs/genetics , Progesterone/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cervix Uteri/pathology , Down-Regulation , Embryo Implantation/genetics , Endometrium/cytology , Endometrium/growth & development , Estrogens/metabolism , Female , Humans , Ki-67 Antigen/metabolism , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/biosynthesis , Mifepristone/pharmacology , Pregnancy , Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , STAT5 Transcription Factor/biosynthesis , Stromal Cells/cytology , Tacrolimus Binding Proteins/genetics , Up-Regulation , Uterus/physiology
11.
J Clin Invest ; 123(9): 4063-75, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23979163

ABSTRACT

There are currently more than 15 million preterm births each year. We propose that gene-environment interaction is a major contributor to preterm birth. To address this experimentally, we generated a mouse model with uterine deletion of Trp53, which exhibits approximately 50% incidence of spontaneous preterm birth due to premature decidual senescence with increased mTORC1 activity and COX2 signaling. Here we provide evidence that this predisposition provoked preterm birth in 100% of females exposed to a mild inflammatory insult with LPS, revealing the high significance of gene-environment interactions in preterm birth. More intriguingly, preterm birth was rescued in LPS-treated Trp53-deficient mice when they were treated with a combination of rapamycin (mTORC1 inhibitor) and progesterone (P4), without adverse effects on maternal or fetal health. These results provide evidence for the cooperative contributions of two sites of action (decidua and ovary) toward preterm birth. Moreover, a similar signature of decidual senescence with increased mTORC1 and COX2 signaling was observed in women undergoing preterm birth. Collectively, our findings show that superimposition of inflammation on genetic predisposition results in high incidence of preterm birth and suggest that combined treatment with low doses of rapamycin and P4 may help reduce the incidence of preterm birth in high-risk women.


Subject(s)
Decidua/metabolism , Gene-Environment Interaction , Premature Birth/prevention & control , 20-Hydroxysteroid Dehydrogenases/metabolism , Animals , Celecoxib , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cytokines/metabolism , Decidua/drug effects , Decidua/immunology , Drug Therapy, Combination , Female , Gene Expression , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Ovary/drug effects , Ovary/immunology , Ovary/pathology , Pregnancy , Premature Birth/genetics , Premature Birth/immunology , Progesterone/pharmacology , Pyrazoles/pharmacology , Receptors, Prolactin/metabolism , Signal Transduction , Sirolimus/pharmacology , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
12.
Mol Endocrinol ; 27(9): 1492-501, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23885093

ABSTRACT

Leukemia inhibitory factor (LIF), a downstream target of estrogen, is essential for implantation in mice. LIF function is thought to be mediated by its binding to LIF receptor (LIFR) and recruitment of coreceptor GP130 (glycoprotein 130), and this receptor complex then activates signal transducer and activator of transcription (STAT)1/3. However, the importance of LIFR and GP130 acting via STAT3 in implantation remains uncertain, because constitutive inactivation of Lifr, Gp130, or Stat3 shows embryonic lethality in mice. To address this issue, we generated mice with conditional deletion of uterine Gp130 or Stat3 and show that both GP130 and STAT3 are critical for uterine receptivity and implantation. Implantation failure in these deleted mice is associated with higher uterine estrogenic responses prior to the time of implantation. These heightened estrogenic responses are not due to changes in ovarian hormone levels or expression of their nuclear receptors. In the deleted mice, estrogen-responsive gene, Lactoferrin (Ltf), and Mucin 1 protein, were up-regulated in the uterus. In addition, progesterone-responsive genes, Hoxa10 and Indian hedgehog (Ihh), were markedly down-regulated in STAT3-inactivated uteri. These changes in uteri of deleted mice were reflected by the failure of differentiation of the luminal epithelium, which is essential for blastocyst attachment.


Subject(s)
Cytokine Receptor gp130/metabolism , Embryo Implantation , Estrogens/metabolism , Gene Deletion , STAT3 Transcription Factor/metabolism , Uterus/metabolism , Animals , Cell Differentiation , Epithelium/metabolism , Epithelium/pathology , Female , Gene Expression Profiling , Infertility, Female/metabolism , Infertility, Female/pathology , Leukemia Inhibitory Factor/metabolism , Mice , Organ Specificity , Ovary/metabolism , Pregnancy , Receptors, OSM-LIF/metabolism
13.
Open Biol ; 3(4): 130035, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23615030

ABSTRACT

Mammalian embryonic diapause is a phenomenon defined by the temporary arrest in blastocyst growth and metabolic activity within the uterus which synchronously becomes quiescent to blastocyst activation and implantation. This reproductive strategy temporally uncouples conception from parturition until environmental or maternal conditions are favourable for the survival of the mother and newborn. The underlying molecular mechanism by which the uterus and embryo temporarily achieve quiescence, maintain blastocyst survival and then resume blastocyst activation with subsequent implantation remains unknown. Here, we show that uterine expression of Msx1 or Msx2, members of an ancient, highly conserved homeobox gene family, persists in three unrelated mammalian species during diapause, followed by rapid downregulation with blastocyst activation and implantation. Mice with uterine inactivation of Msx1 and Msx2 fail to achieve diapause and reactivation. Remarkably, the North American mink and Australian tammar wallaby share similar expression patterns of MSX1 or MSX2 as in mice-it persists during diapause and is rapidly downregulated upon blastocyst activation and implantation. Evidence from mouse studies suggests that the effects of Msx genes in diapause are mediated through Wnt5a, a known transcriptional target of uterine Msx. These studies provide strong evidence that the Msx gene family constitutes a common conserved molecular mediator in the uterus during embryonic diapause to improve female reproductive fitness.


Subject(s)
Homeodomain Proteins/metabolism , MSX1 Transcription Factor/metabolism , Animals , Blastocyst/cytology , Blastocyst/metabolism , Down-Regulation , Embryo Implantation , Embryonic Development , Female , Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Mice , Mink/metabolism , Uterus/metabolism , Wnt Proteins/metabolism , Wnt-5a Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...