Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 1174, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36859534

ABSTRACT

Placental abnormalities have been sporadically implicated as a source of developmental heart defects. Yet it remains unknown how often the placenta is at the root of congenital heart defects (CHDs), and what the cellular mechanisms are that underpin this connection. Here, we selected three mouse mutant lines, Atp11a, Smg9 and Ssr2, that presented with placental and heart defects in a recent phenotyping screen, resulting in embryonic lethality. To dissect phenotype causality, we generated embryo- and trophoblast-specific conditional knockouts for each of these lines. This was facilitated by the establishment of a new transgenic mouse, Sox2-Flp, that enables the efficient generation of trophoblast-specific conditional knockouts. We demonstrate a strictly trophoblast-driven cause of the CHD and embryonic lethality in one of the three lines (Atp11a) and a significant contribution of the placenta to the embryonic phenotypes in another line (Smg9). Importantly, our data reveal defects in the maternal blood-facing syncytiotrophoblast layer as a shared pathology in placentally induced CHD models. This study highlights the placenta as a significant source of developmental heart disorders, insights that will transform our understanding of the vast number of unexplained congenital heart defects.


Subject(s)
Heart Diseases , Trophoblasts , Female , Pregnancy , Animals , Mice , Placenta , Heart , Epithelial Cells , Mice, Transgenic
2.
Mol Cell Biol ; 38(9)2018 05 01.
Article in English | MEDLINE | ID: mdl-29463649

ABSTRACT

The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) is an essential regulator of placental development. To gain deeper insights into placental PPARγ signaling, we dissected its regulation of the Muc1 promoter. We find that, unlike prototypic target activation by heterodimeric receptors, which is either stimulated by or refractory to retinoid X receptor (RXR) ligands (rexinoids), the induction of Muc1 by liganded PPARγ requires RXRα but is inhibited by rexinoids. We demonstrate that this inhibition is mediated by the activation function 2 (AF2) domain of RXRα and that Muc1 activation entails altered AF2 structures of both PPARγ and RXRα. This unique regulation of Muc1 reflects specific coactivation of PPARγ-RXRα heterodimers by the transcription cofactor ligand-dependent corepressor (LCoR), corroborated by significant downregulation of Muc1 in Lcor-null placentas. LCoR interacts with PPARγ and RXRα in a synergistic fashion via adjacent noncanonical protein motifs, and the AF2 domain of ligand-bound RXRα inhibits this interaction. We further identify the transcription factor Krüppel-like factor 6 (KLF6) as a critical regulator of placental development and a component of Muc1 regulation in cooperation with PPARγ, RXRα, and LCoR. Combined, these studies reveal new principles and players in nuclear receptor function in general and placental PPARγ signaling in particular.


Subject(s)
Mucin-1/metabolism , PPAR gamma/metabolism , Repressor Proteins/metabolism , Animals , Carrier Proteins , Cell Line , Co-Repressor Proteins , Female , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Kruppel-Like Factor 6/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mucin-1/genetics , PPAR gamma/physiology , Pregnancy , Promoter Regions, Genetic/genetics , Repressor Proteins/genetics , Retinoid X Receptor alpha/metabolism , Retinoid X Receptors/metabolism , Transcriptional Activation
3.
Dev Biol ; 372(1): 143-55, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22967998

ABSTRACT

The nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) is essential for placental development. For insights into its functions in the placenta, we screened for PPARγ-regulated genes by integrating expression profiles of Pparg-null and Rxra-null placentas with those of WT and Pparg-null trophoblast stem cells differentiated in the presence or absence of a PPARγ agonist. Intersection of these paradigms identified high-probability PPARγ target genes. A few of these genes were previously reported as PPARγ targets in other tissues, but most are new in the context of either PPARγ or placental biology. Transcriptional profiling demonstrated a widespread role for the coactivator NCOA6/AIB3, but not MED1/PBP, in PPARγ-dependent placental gene expression. Spatial and temporal expression analyses revealed that PPARγ impacts genes in diverse trophoblast lineages and during different stages of differentiation. We further validated the Ldhb gene, which encodes the H isoform of lactate dehydrogenase, as a robust PPARγ target in trophoblasts, and propose a hypothetical model that integrates it with a network of PPARγ-regulated genes into a novel pathway of placental fuel metabolism. These findings offer insights not only into the placental functions of PPARγ, but also into unique, previously unsuspected biosynthetic functions of trophoblasts.


Subject(s)
PPAR gamma/genetics , Placentation/genetics , Animals , Chromatin Immunoprecipitation , Female , Gene Expression Regulation, Developmental , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Male , Metabolic Networks and Pathways/genetics , Mice , Mice, Transgenic , PPAR gamma/metabolism , Placenta/metabolism , Pregnancy , Stem Cells/cytology , Stem Cells/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism
4.
J Biol Chem ; 287(12): 8662-74, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22277651

ABSTRACT

The widely expressed transcriptional coregulator, ligand-dependent corepressor (LCoR), initially characterized as a regulator of nuclear receptor-mediated transactivation, functions through recruitment of C-terminal binding proteins (CtBPs) and histone deacetylases (HDACs) to its N-terminal and central domains, respectively. We performed a yeast two-hybrid screen for novel cofactors, and identified an interaction between the C-terminal domain of LCoR and the transcription factor Krüppel-like factor 6 (KLF6), a putative tumor suppressor in prostate cancer. Subsequent experiments revealed LCoR regulation of several KLF6 target genes notably p21(WAF1/CIP1) (CDKN1A) and to a lesser extent E-cadherin (CDH1), indicating that LCoR regulates gene transcription through multiple classes of transcription factors. In multiple cancer cells, LCoR and KLF6 bind together on the promoters of the genes encoding CDKN1A and CDH1. LCoR contributes to KLF6-mediated transcriptional repression in a promoter- and cell type-dependent manner. Its inhibition of reporter constructs driven by the CDKN1A and CDH1 promoters in PC-3 prostate carcinoma cells is sensitive to treatment with the HDAC inhibitor trichostatin A. Additionally, the LCoR cofactor CtBP1 bound the same promoters and augmented the LCoR-dependent repression in PC-3 cells. Consistent with their inferred roles in transcriptional repression, siRNA-mediated knockdown of KLF6, LCoR, or CtBP1 in PC-3 cells induced expression of CDKN1A and CDH1 and additional KLF6 target genes. We propose a novel model of LCoR function in which promoter-bound KLF6 inhibits transcription of the CDKN1A gene and other genes as well by tethering a transcriptional corepressor complex containing LCoR, with specific contributions by CtBP1 and HDACs.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation , Kruppel-Like Transcription Factors/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/genetics , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics
5.
PPAR Res ; 2008: 142082, 2008.
Article in English | MEDLINE | ID: mdl-18288278

ABSTRACT

With the major attention to the pivotal roles of PPARs in diverse aspects of energy metabolism, the essential functions of PPARgamma and PPARbeta/delta in placental development came as a surprise and were often considered a nuisance en route to their genetic analysis. However, these findings provided an opportune entrée into placental biology. Genetic and pharmacological studies, primarily of knockout animal models and cell culture, uncovered networks of PPARgamma and PPARdelta, their heterodimeric RXR partners, associated transcriptional coactivators, and target genes, that regulate various aspects of placental development and function. These studies furnish both specific information about trophoblasts and the placenta and potential hints about the functions of PPARs in other tissues and cell types. They reveal that the remarkable versatility of PPARs extends beyond the orchestration of metabolism to the regulation of cellular differentiation, tissue development, and trophoblast-specific functions. This information and its implications are the subject of this review.

6.
Proc Natl Acad Sci U S A ; 104(42): 16627-32, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17921248

ABSTRACT

Lipodystrophies are syndromes of adipose tissue degeneration associated with severe defects in lipid and glucose homeostasis. We report here the generation and analysis of Pparg(ldi), a targeted allele that confers conditional dominant lipodystrophy in mice. The Pparg(ldi) allele was generated by insertion of the Tet activator (tTA) and a tTA-regulated Flag-Pparg1 transgene into the Pparg gene. Unexpectedly, tTA elicits mild lipodystrophy, insulin resistance, and dyslipidemia, and the Flag-PPARgamma1 transgene surprisingly exacerbates these traits. Doxycycline can both completely prevent and reverse these phenotypes, providing a mouse model of inducible lipodystrophy. Embryonic fibroblasts from either Pparg(ldi/+) or the phenotypically similar aP2-nSrebp1c (Sr) transgenic mice undergo robust adipogenesis, suggesting that neither strain develops lipodystrophy because of defective adipocyte differentiation. In addition, Pparg(ldi/+) adipose tissue shares extensive gene expression aberrations with that of Sr mice, authenticating the phenotype at the molecular level and revealing a common expression signature of lipodystrophic fat. Thus, the Pparg(ldi/+) mouse provides a conditional animal model for studying lipodystrophy and its associated physiology and gene expression.


Subject(s)
Disease Models, Animal , Lipodystrophy/genetics , Mice, Transgenic , PPAR gamma/genetics , Adipogenesis/genetics , Alleles , Animals , Doxycycline/pharmacology , Fibroblasts/metabolism , Gene Expression , Insulin Resistance/genetics , Lipodystrophy/pathology , Mice , Promoter Regions, Genetic/drug effects , Sterol Regulatory Element Binding Protein 1/genetics , Tetracycline/pharmacology , Trans-Activators/genetics
7.
Mol Cell Biol ; 24(24): 10661-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15572671

ABSTRACT

The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) is essential for placental development. Here, we show that the mucin gene Muc1 is a PPARgamma target, whose expression is lost in PPARgamma null placentas. During differentiation of trophoblast stem cells, PPARgamma is strongly induced, and Muc1 expression is upregulated by the PPARgamma agonist rosiglitazone. Muc1 promoter is activated strongly and specifically by liganded PPARgamma but not PPARalpha or PPARdelta. A PPAR binding site (DR1) in the proximal Muc1 promoter acts as a basal silencer in the absence of PPARgamma, and its cooperation with a composite upstream enhancer element is both necessary and sufficient for PPARgamma-dependent induction of Muc1. In the placenta, MUC1 protein is localized exclusively to the apical surface of the labyrinthine trophoblast around maternal blood sinuses, resembling its luminal localization on secretory epithelia. Last, variably penetrant maternal blood sinus dilation in Muc1-deficient placentas suggests that Muc1 regulation by PPARgamma contributes to normal placental development but also that the essential functions of PPARgamma in the organ are mediated by other targets.


Subject(s)
Gene Expression Regulation, Developmental , Mucin-1/genetics , PPAR gamma/metabolism , Transcription, Genetic , Trophoblasts/metabolism , Animals , Binding Sites , Cells, Cultured , Crosses, Genetic , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic , Female , Fluorescent Dyes , Genes, Reporter , Hypoglycemic Agents/pharmacology , Ligands , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , PPAR gamma/agonists , Pregnancy , Promoter Regions, Genetic , Rosiglitazone , Stem Cells/cytology , Thiazolidinediones/pharmacology , Trophoblasts/cytology , Up-Regulation
8.
J Biol Chem ; 277(43): 40659-65, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12189148

ABSTRACT

Chemoattractant-stimulated phagocytes increase their glucose uptake and divert energy production from glycolysis to the pentose phosphate pathway to generate NADPH. NADPH is a required cofactor for the NADPH oxidase to produce reactive oxygen metabolites, an important microbicidal tool in host defense. p21-Activated kinases (Paks) are regulated by the GTPases Rac and Cdc42 and control actin dynamics and phosphorylation of the oxidase component p47(phox). Here we report the interaction of Pak with phosphoglycerate mutase (PGAM)-B, an enzyme of the glycolytic pathway. Activated Pak1 inhibits glycolysis by association of its catalytic domain with PGAM-B and subsequent phosphorylation of the enzyme on serine residues 23 and 118, thereby abolishing PGAM activity. Leukocyte activation through chemoattractant receptors leads to Pak activation and transient inhibition of endogenous PGAM-B activity. Consistent with these observations, treatment of neutrophils with phosphoglycolic acid, a competitive PGAM-B inhibitor, increases upstream intermediates, thereby amplifying the respiratory burst. These results demonstrate that Rho GTPases regulate the glycolytic pathway through Pak and suggest a link between chemoattractant signaling and metabolic responses to enhance host defense.


Subject(s)
NADPH Oxidases/metabolism , Phagocytes/enzymology , Protein Serine-Threonine Kinases/metabolism , Up-Regulation , Humans , Phosphoglycerate Mutase/antagonists & inhibitors , Phosphoglycerate Mutase/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...