Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Am J Physiol Renal Physiol ; 321(6): F705-F714, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34632813

ABSTRACT

The epithelial Na+ channel (ENaC) promotes the absorption of Na+ in the aldosterone-sensitive distal nephron, colon, and respiratory epithelia. Deletion of genes encoding subunits of ENaC results in early postnatal mortality. Here, we present the initial characterization of a mouse with dramatically suppressed expression of the ENaC γ-subunit. We used this hypomorphic (γmt) allele to explore the importance of this subunit in homeostasis of electrolytes and body fluid volume. At baseline, γ-subunit expression in γmt/mt mice was markedly suppressed in the kidney and lung, whereas electrolytes resembled those of littermate controls. Aldosterone levels in γmt/mt mice exceeded those seen in littermate controls. Quantitative magnetic resonance measurement of body composition revealed similar baseline body water, lean tissue mass, and fat tissue mass in γmt/mt mice and controls. γmt/mt mice exhibited a more rapid decline in body water and lean tissue mass in response to a low-Na+ diet than the controls. Replacement of drinking water with 2% saline selectively and transiently increased body water and lean tissue mass in γmt/mt mice relative to the controls. Lower blood pressures were variably observed in γmt/mt mice on a high-salt diet compared with the controls. γmt/mt also exhibited reduced diurnal blood pressure variation, a "nondipping" phenotype, on a high-Na+ diet. Although ENaC in the renal tubules and colon works to prevent extracellular fluid volume depletion, our observations suggest that ENaC in other tissues may participate in regulating extracellular fluid volume and blood pressure.NEW & NOTEWORTHY A mouse with globally suppressed expression of the epithelial Na+ channel γ-subunit showed enhanced sensitivity to dietary salt, including a transient increase in total body fluid, reduced blood pressure, and reduced diurnal blood pressure variation when given a dietary NaCl challenge. These results point to a role for the epithelial Na+ channel in regulating body fluid and blood pressure beyond classical transepithelial Na+ transport mechanisms.


Subject(s)
Blood Pressure , Blood Volume , Diet, Sodium-Restricted , Epithelial Sodium Channels/deficiency , Kidney/metabolism , Lung/metabolism , Sodium Chloride, Dietary/metabolism , Water-Electrolyte Balance , Animals , Biomarkers/blood , Biomarkers/urine , Body Composition , Epithelial Sodium Channels/genetics , Female , Male , Mice, Knockout , Organism Hydration Status , Sodium Chloride, Dietary/administration & dosage , Sodium Chloride, Dietary/toxicity
2.
Am J Obstet Gynecol ; 223(3): 312-321, 2020 09.
Article in English | MEDLINE | ID: mdl-32565236

ABSTRACT

Recent revolutionary advances at the intersection of medicine, omics, data sciences, computing, epidemiology, and related technologies inspire us to ponder their impact on health. Their potential impact is particularly germane to the biology of pregnancy and perinatal medicine, where limited improvement in health outcomes for women and children has remained a global challenge. We assembled a group of experts to establish a Pregnancy Think Tank to discuss a broad spectrum of major gestational disorders and adverse pregnancy outcomes that affect maternal-infant lifelong health and should serve as targets for leveraging the many recent advances. This report reflects avenues for future effects that hold great potential in 3 major areas: developmental genomics, including the application of methodologies designed to bridge genotypes, physiology, and diseases, addressing vexing questions in early human development; gestational physiology, from immune tolerance to growth and the timing of parturition; and personalized and population medicine, focusing on amalgamating health record data and deep phenotypes to create broad knowledge that can be integrated into healthcare systems and drive discovery to address pregnancy-related disease and promote general health. We propose a series of questions reflecting development, systems biology, diseases, clinical approaches and tools, and population health, and a call for scientific action. Clearly, transdisciplinary science must advance and accelerate to address adverse pregnancy outcomes. Disciplines not traditionally involved in the reproductive sciences, such as computer science, engineering, mathematics, and pharmacology, should be engaged at the study design phase to optimize the information gathered and to identify and further evaluate potentially actionable therapeutic targets. Information sources should include noninvasive personalized sensors and monitors, alongside instructive "liquid biopsies" for noninvasive pregnancy assessment. Future research should also address the diversity of human cohorts in terms of geography, racial and ethnic distributions, and social and health disparities. Modern technologies, for both data-gathering and data-analyzing, make this possible at a scale that was previously unachievable. Finally, the psychosocial and economic environment in which pregnancy takes place must be considered to promote the health and wellness of communities worldwide.


Subject(s)
Health Promotion/trends , Pregnancy Outcome , Economics , Female , Fetal Development/genetics , Fetal Development/physiology , Humans , Perinatal Care , Pregnancy , Pregnancy Complications/ethnology , Pregnancy Complications/genetics , Pregnancy Complications/physiopathology , Pregnancy Outcome/epidemiology , Pregnancy Outcome/genetics , Psychology
3.
Pediatr Cardiol ; 41(1): 220, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31680222

ABSTRACT

The original version of this article unfortunately contained a mistake. In reviewing the phenotype associated with Mapk14 (p38alpha MAPK) mutation as evaluated by Adams et al. (2000) using tetraploid aggregation chimeric embryos, the authors mistakenly stated that rescue of embryo lethality was short-lived and that embryos died two days later of non-placenta-related causes. In fact, as reported by Adams et al. (2000), when the placental defect of global null embryos was rescued, p38alpha(-/-) embryos developed to term and were normal in appearance. The authors apologize for the error.

4.
Immunity ; 33(5): 699-712, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21093321

ABSTRACT

Peroxisome proliferator-activated receptor γ (PPARγ) is a lipid-activated transcription factor regulating lipid metabolism and inflammatory response in macrophages and dendritic cells (DCs). These immune cells exposed to distinct inflammatory milieu show cell type specification as a result of altered gene expression. We demonstrate here a mechanism how inflammatory molecules modulate PPARγ signaling in distinct subsets of cells. Proinflammatory molecules inhibited whereas interleukin-4 (IL-4) stimulated PPARγ activity in macrophages and DCs. Furthermore, IL-4 signaling augmented PPARγ activity through an interaction between PPARγ and signal transducer and activators of transcription 6 (STAT6) on promoters of PPARγ target genes, including FABP4. Thus, STAT6 acts as a facilitating factor for PPARγ by promoting DNA binding and consequently increasing the number of regulated genes and the magnitude of responses. This interaction, underpinning cell type-specific responses, represents a unique way of controlling nuclear receptor signaling by inflammatory molecules in immune cells.


Subject(s)
Dendritic Cells/metabolism , Gene Expression Regulation , Macrophages/metabolism , PPAR gamma/metabolism , STAT6 Transcription Factor/metabolism , Animals , Fatty Acid-Binding Proteins/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin-4/metabolism , Mice , Promoter Regions, Genetic
5.
Pediatr Cardiol ; 40(7): 1359-1366, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31342113

ABSTRACT

If viewed as a movie, heart morphogenesis appears to unfold in a continuous and seamless manner. At the mechanistic level, however, a series of discreet and separable processes sequentially underlie heart development. This is evident in examining the expansion of the ventricular wall, which accounts for most of the contractile force of each heartbeat. Ventricular wall expansion is driven by cardiomyocyte proliferation coupled with a morphogenetic program that causes wall thickening rather than lengthening. Although most studies of these processes have focused on heart-intrinsic processes, it is increasingly clear that extracardiac events influence or even direct heart morphogenesis. In this review, we specifically consider mechanisms responsible for coordinating cardiomyocyte proliferation and ventricular wall expansion in mammalian development, relying primarily on studies from mouse development where a wealth of molecular and genetic data have been accumulated.


Subject(s)
Cell Proliferation , Heart Ventricles/embryology , Morphogenesis/physiology , Myocytes, Cardiac/metabolism , Animals , Heart Ventricles/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Mice
6.
Genesis ; 54(1): 53-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26663459

ABSTRACT

Among transgenic mice with ubiquitous Cre recombinase activity, all strains to date excise loxP-flanked (floxed) alleles either at or before the zygote stage or at nondescript stages of development. This manuscript describes a new mouse strain, in which Cre recombinase, integrated into the Esrrb locus, efficiently excises floxed alleles in pre-implantation embryos at the onset of the four-cell stage. By enabling inactivation of genes only after the embryo has undergone two cleavages, this strain should facilitate in vivo studies of genes with essential gametic or zygotic functions. In addition, this study describes a new, highly pluripotent hybrid C57BL/6J x 129S1/SvImJ mouse embryonic stem cell line, HYB12, in which this knockin and additional targeted alleles have been generated.


Subject(s)
Alleles , Receptors, Estrogen/genetics , Animals , Cell Line , Gene Deletion , Gene Transfer Techniques , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombination, Genetic , Viral Proteins/genetics , Zygote/metabolism
7.
Clin Immunol ; 163: 91-5, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26769277

ABSTRACT

Severe combined immune deficiency (SCID) is a group of genetically heterogeneous diseases caused by an early block in T cell differentiation and present with life threatening infections, often within the first year of life. Janus kinase (JAK)3 gene mutations have been found to cause autosomal recessive T-B+ SCID phenotype. In this study we describe three patients with a novel deep intronic mis-splicing mutation in JAK3 as a cause of T-B+NK- SCID highlighting the need for careful evaluation of intronic regulatory elements of known genes associated with clearly defined clinical phenotypes. We present the cases and discuss the current literature.


Subject(s)
B-Lymphocytes/immunology , Introns/genetics , Janus Kinase 3/genetics , Killer Cells, Natural/immunology , RNA Splicing/genetics , Severe Combined Immunodeficiency/genetics , T-Lymphocytes/immunology , Consanguinity , Female , Humans , Infant , Janus Kinase 3/immunology , Male , Mutation , Phenotype , Severe Combined Immunodeficiency/immunology
8.
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
9.
J Hepatol ; 59(5): 1045-53, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23831119

ABSTRACT

BACKGROUND & AIMS: PPARγ plays an essential role in the transcriptional regulation of genes involved in lipid and glucose metabolism, insulin sensitivity, and inflammation. We recently demonstrated that PPARγ plays a causative role in hepatocyte lipid deposition, contributing to the pathogenesis of hepatic steatosis. In this study, we investigated the role of PPARγ in the inflammatory and fibrogenic response of the liver. METHODS: Heterozygous floxed/null Cre/LoxP mice with targeted deletion of PPARγ in either hepatocytes (Alb-Cre), macrophages (LysM-Cre) or hepatic stellate cells (HSCs) (aP2-Cre) were submitted to carbon tetrachloride (CCl4) liver injury. Further analyses were performed in precision-cut liver slices (PCLS) and primary cultures of hepatocytes, macrophages, and HSCs. RESULTS: LysM-Cre mice displayed an exacerbated response to chronic CCl4 injury and showed higher necroinflammatory injury, lipid peroxidation, inflammatory infiltrate, cleaved-caspase-3 and caspase 3/7 activity, and COX-2, TNF-α, CXCL2, and IL-1ß expression than Alb-Cre and control mice. The deleterious effects of PPARγ disruption in liver macrophages were confirmed in an acute model of CCl4 injury as well as in PCLS incubated with LPS. Moreover, LysM-Cre mice showed an aggravated fibrogenic response to CCl4, as revealed by more prominent Sirius Red and Masson's trichrome staining, elevated hydroxyproline content and induced α-SMA and TIMP-1 expression. Importantly, aP2-Cre mice with specific disruption of PPARγ in HSCs, as confirmed by immunocytochemical analysis of individual liver cells, also showed exacerbated liver damage and fibrogenic response to CCl4. CONCLUSIONS: These data unveil anti-inflammatory and anti-fibrogenic roles for PPARγ in non-parenchymal liver cells.


Subject(s)
Hepatic Stellate Cells/pathology , Hepatocytes/pathology , Inflammation/physiopathology , Liver Cirrhosis/physiopathology , Macrophages/pathology , PPAR gamma/deficiency , PPAR gamma/physiology , Actins/metabolism , Animals , Carbon Tetrachloride/adverse effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , PPAR gamma/genetics , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Tissue Inhibitor of Metalloproteinase-1/metabolism
10.
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
11.
Stem Cells ; 29(7): 1034-40, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21544899

ABSTRACT

Adipose tissue is the primary energy reservoir in the body and an important endocrine organ that plays roles in energy homeostasis, feeding, insulin sensitivity, and inflammation. While it was tacitly assumed that fat in different anatomical locations had a common origin and homogenous function, it is now clear that regional differences exist in adipose tissue characteristics and function. This is exemplified by the link between increased deep abdominal or visceral fat, but not peripheral adipose tissue and the metabolic disturbances associated with obesity. Regional differences in fat function are due in large part to distinct adipocyte populations that comprise the different fat depots. Evidence accrued primarily in the last decade indicates that the distinct adipocyte populations are generated by a number of processes during and after development. These include the production of adipocytes from different germ cell layers, the formation of distinct preadipocyte populations from mesenchymal progenitors of mesodermal origin, and the production of adipocytes from hematopoietic stem cells from the bone marrow. This review will examine each of these process and their relevance to normal adipose tissue formation and contribution to obesity-related diseases.


Subject(s)
Adipocytes/cytology , Adipose Tissue/cytology , Intra-Abdominal Fat/cytology , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Humans , Intra-Abdominal Fat/metabolism , Mice
12.
FASEB J ; 25(8): 2538-50, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21507897

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) γ is a nuclear receptor central to glucose and lipid homeostasis. PPARγ role in nonalcoholic fatty liver disease is controversial because PPARγ overexpression is a general property of steatotic livers, but its activation by thiazolidinediones reduces hepatic steatosis. Here, we investigated hepatic PPARγ function by using Cre-loxP technology to generate hepatocyte (PPARγ(Δhep))- and macrophage (PPARγ(Δmac))-specific PPARγ-knockout mice. Targeted deletion of PPARγ in hepatocytes, and to a lesser extent in macrophages, protected mice against high-fat diet-induced hepatic steatosis. Down-regulated expression of genes involved in lipogenesis (SCD1, SREBP-1c, and ACC), lipid transport (CD36/FAT, L-FABP, and MTP), and ß-oxidation (PPARα and ACO) was observed in PPARγ(Δhep) mice. Moreover, PPARγ(Δhep) mice showed improved glucose tolerance and reduced PEPCK expression without changes in Pcx, Fbp1, and G6Pc expression and CREB and JNK phosphorylation. In precision-cut liver slices (PCLSs) and hepatocytes, rosiglitazone either alone or in combination with oleic acid increased triglyceride accumulation, an effect that was blocked by the PPARγ antagonist biphenol A diglycidyl ether (BADGE). PCLSs and hepatocytes from PPARγ(Δhep) mice showed blunted responses to rosiglitazone and oleic acid, whereas the response to these compounds remained intact in PCLSs from PPARγ(Δmac) mice. Collectively, these findings establish PPARγ expression in hepatocytes as a prosteatotic factor in fatty liver disease.


Subject(s)
Fatty Liver/etiology , Fatty Liver/physiopathology , Obesity/complications , Obesity/physiopathology , PPAR gamma/physiology , Animals , Base Sequence , DNA Primers/genetics , Dietary Fats/administration & dosage , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression , Gene Targeting , Glucose/metabolism , Hepatocytes/physiology , Kupffer Cells/physiology , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease , Obesity/genetics , Obesity/pathology , Organ Specificity , PPAR gamma/deficiency , PPAR gamma/genetics
13.
Nat Med ; 9(12): 1491-7, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625542

ABSTRACT

Thiazolidinediones (TZDs) are insulin-sensitizing drugs and are potent agonists of the nuclear peroxisome proliferator-activated receptor-gamma (PPAR-gamma). Although muscle is the major organ responsible for insulin-stimulated glucose disposal, PPAR-gamma is more highly expressed in adipose tissue than in muscle. To address this issue, we used the Cre-loxP system to knock out Pparg, the gene encoding PPAR-gamma, in mouse skeletal muscle. As early as 4 months of age, mice with targeted disruption of PPAR-gamma in muscle showed glucose intolerance and progressive insulin resistance. Using the hyperinsulinemic-euglycemic clamp technique, the in vivo insulin-stimulated glucose disposal rate (IS-GDR) was reduced by approximately 80% and was unchanged by 3 weeks of TZD treatment. These effects reveal a crucial role for muscle PPAR-gamma in the maintenance of skeletal muscle insulin action, the etiology of insulin resistance and the action of TZDs.


Subject(s)
Insulin Resistance/physiology , Muscle Proteins , Muscle, Skeletal/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Gene Expression/drug effects , Glucose Clamp Technique , Glucose Transporter Type 4 , Insulin/metabolism , Insulin Resistance/genetics , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Signal Transduction , Thiazolidinediones/pharmacology , Transcription Factors/agonists
14.
FASEB J ; 23(9): 2968-77, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19395477

ABSTRACT

Ligands of peroxisome proliferator-activated receptor-gamma (PPAR-gamma) abrogate the stimulation of collagen gene transcription induced by transforming growth factor-beta (TGF-beta). Here, we delineate the mechanisms underlying this important novel physiological function for PPAR-gamma in connective tissue homeostasis. First, we demonstrated that antagonistic regulation of TGF-beta activity by PPAR-gamma ligands involves cellular PPAR-gamma, since 15-deoxy-Delta12,14-prostaglandin J(2) (15d-PGJ(2)) failed to block TGF-beta-induced responses in either primary cultures of PPAR-gamma-null murine embryonic fibroblasts, or in normal human skin fibroblasts with RNAi-mediated knockdown of PPAR-gamma. Next, we examined the molecular basis underlying the abrogation of TGF-beta signaling by PPAR-gamma in normal human fibroblasts in culture. The results demonstrated that Smad-dependent transcriptional responses were blocked by PPAR-gamma without preventing Smad2/3 activation. In contrast, the interaction between activated Smad2/3 and the transcriptional coactivator and histone acetyltransferase p300 induced by TGF-beta, and the accumulation of p300 on consensus Smad-binding DNA sequences and histone H4 hyperacetylation at the COL1A2 locus, were all prevented by PPAR-gamma. Wild-type p300, but not a mutant form of p300 lacking functional histone acetyltransferase, was able to restore TGF-beta-induced stimulation of COL1A2 in the presence of PPAR-gamma ligands. Collectively, these results indicate that PPAR-gamma blocked Smad-mediated transcriptional responses by preventing p300 recruitment and histone H4 hyperacetylation, resulting in the inhibition of TGF-beta-induced collagen gene expression. Pharmacological activation of PPAR-gamma thus may represent a novel therapeutic approach to target p300-dependent TGF-beta profibrotic responses such as stimulation of collagen gene expression.


Subject(s)
Collagen/genetics , PPAR gamma/physiology , Smad Proteins/physiology , p300-CBP Transcription Factors/metabolism , Acetylation , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Histones/metabolism , Humans , Mice , Protein Transport , Transcriptional Activation , Transforming Growth Factor beta/physiology , p300-CBP Transcription Factors/genetics
15.
Placenta ; 88: 61-67, 2019 12.
Article in English | MEDLINE | ID: mdl-31675530

ABSTRACT

INTRODUCTION: Imprinted genes are preferentially expressed from one parentally inherited allele, and many are crucial to the regulation of placental function and fetal growth. Murine Krüppel-like factor 14 (Klf14) is a maternally expressed imprinted transcription factor that is a component of the Mest imprinted gene cluster on mouse chromosome 6. We sought to determine if loss of Klf14 expression alters the course of normal mouse extraembryonic development. We also used high-throughput RNA sequencing (RNAseq) to identify a set of differentially expressed genes (DEGs) in placentas with loss of Klf14. METHODS: We generated a Klf14 knockout (Klf14null) mouse using recombineering and transgenic approaches. To identify DEGs in the mouse placenta we compared mRNA transcriptomes derived from 17.5dpc Klf14matKO and wild-type littermate placentas by RNAseq. Candidate DEGs were confirmed with quantitative reverse transcription PCR (qPCR) on an independent cohort of male and female gestational age matched Klf14matKO placentas. RESULTS: We found that 17.5dpc placentas inheriting a maternal null allele (Klf14matKO) had a modest overgrowth phenotype and a near complete ablation of Klf14 expression. However, there was no effect on fetal growth. We identified 20 DEGs differentially expressed in Klf14matKO placentas by RNAseq, and subsequently validated five that are highly upregulated (Begain, Col26a1, Fbln5, Gdf10, and Nell1) by qPCR. The most enriched functional gene-networks included those classified as regulating cellular development and metabolism. CONCLUSION: These results suggest that loss of the maternal Klf14 locus in the mouse placenta acts results in changes in gene expression patterns that modulate placental growth.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Placentation , Animals , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Female , Gene Regulatory Networks , Genomic Imprinting , Growth Differentiation Factor 10/metabolism , Mice, Knockout , Pregnancy , Recombinant Proteins/metabolism
16.
Endocrinology ; 159(12): 3937-3949, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30351430

ABSTRACT

Trophoblast hypoxia and injury, key components of placental dysfunction, are associated with fetal growth restriction and other complications of pregnancy. Accumulation of lipid droplets has been found in hypoxic nonplacental cells. Unique to pregnancy, lipid accumulation in the placenta might perturb lipid transport to the fetus. We tested the hypothesis that hypoxia leads to accumulation of lipid droplets in human trophoblasts and that trophoblastic PLIN proteins play a key role in this process. We found that hypoxia promotes the accumulation of lipid droplets in primary human trophoblasts. A similar accretion of lipid droplets was found in placental villi in vivo from pregnancies complicated by fetal growth restriction. In both situations, these changes were associated with an increased level of cellular triglycerides. Exposure of trophoblasts to hypoxia led to reduced fatty acid efflux and oxidation with no change in fatty acid uptake or synthesis. We further found that hypoxia markedly stimulated PLIN2 mRNA synthesis and protein expression, which colocalized to lipid droplets. Knockdown of PLIN2, but not PLIN3, enhanced trophoblast apoptotic death, and overexpression of PLIN2 promoted cell viability. Collectively, our data indicate that hypoxia enhances trophoblastic lipid retention in the form of lipid droplets and that PLIN2 plays a key role in this process and in trophoblast defense against apoptotic death. These findings also imply that this protective mechanism may lead to diminished trafficking of lipids to the developing fetus.


Subject(s)
Hypoxia/genetics , Hypoxia/metabolism , Lipid Droplets/metabolism , Lipid Metabolism/genetics , Perilipin-2/physiology , Trophoblasts/metabolism , Cell Survival/genetics , Cells, Cultured , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Humans , Hypoxia/pathology , Infant, Newborn , Placenta/metabolism , Placenta/pathology , Pregnancy , Trophoblasts/pathology
17.
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
18.
Endocrinology ; 148(8): 3625-34, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17463056

ABSTRACT

The nuclear receptor peroxisome proliferator activated receptor gamma (PPARgamma) is essential for murine placental development. We previously showed that activation of PPARgamma in primary human trophoblasts enhances the uptake of fatty acids and alters the expression of several proteins associated with fatty acid trafficking. In this study we examined the effect of ligand-activated PPARgamma on placental development and transplacental fatty acid transport in wild-type (wt) and PPARgamma(+/-) embryos. We found that exposure of pregnant mice to the PPARgamma agonist rosiglitazone for 8 d (embryonic d 10.5-18.5) reduced the weights of wt, but not PPARgamma(+/-) placentas and embryos. Exposure to rosiglitazone reduced the thickness of the spongiotrophoblast layer and the surface area of labyrinthine vasculature, and altered expression of proteins implicated in placental development. The expression of fatty acid transport protein 1 (FATP1), FATP4, adipose differentiation related protein, S3-12, and myocardial lipid droplet protein was enhanced in placentas of rosiglitazone-treated wt embryos, whereas the expression of FATP-2, -3, and -6 was decreased. Additionally, rosiglitazone treatment was associated with enhanced accumulation of the fatty acid analog 15-(p-iodophenyl)-3-(R, S)-methyl pentadecanoic acid in the placenta, but not in the embryos. These results demonstrate that in vivo activation of PPARgamma modulates placental morphology and fatty acid accumulation.


Subject(s)
Fatty Acids/pharmacokinetics , PPAR gamma/genetics , PPAR gamma/metabolism , Placenta/cytology , Placenta/metabolism , Animals , Female , Fetal Weight/drug effects , Fetal Weight/physiology , Genotype , Hypoglycemic Agents/pharmacology , Iodine Radioisotopes , Iodobenzenes/pharmacokinetics , Ligands , Male , Maternal-Fetal Exchange/drug effects , Maternal-Fetal Exchange/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Placenta/drug effects , Pregnancy , Rosiglitazone , Thiazolidinediones/pharmacology
19.
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
20.
Mol Cell Endocrinol ; 249(1-2): 10-5, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16574314

ABSTRACT

At different stages of placental development the cytotrophoblasts differentiate into specialized cells that are vital for specific placental tasks. These types include the invasive trophoblasts, which are responsible for invasion of the placenta into the uterine wall, and syncytiotrophoblasts, which form a barrier between the maternal and fetal circulations, govern trans-placental transport of gas, nutrient and waste, and produce placental hormones. Recent research illuminated the role of the nuclear receptor peroxisome proliferator-activated receptor-gamma (PPAR gamma) in the areas of adipocyte and macrophage biology, insulin action, bioenergetics and inflammation. It was somewhat surprising that PPAR gamma was also found to play a pivotal role in placental biology. In this review we summarize recent data, which show that PPAR gamma is expressed in the placenta, particularly in trophoblasts, and is essential for placental development, trophoblast invasion, differentiation of cytotrophoblasts into syncytium, and regulation of fat accumulation in trophoblasts. PPAR gamma may also play a role in modulating fetal membrane signals toward parturition. The data presented here underscore the need for a focused investigation of the unique aspects of PPAR gamma function in trophoblasts, which may have direct implications for the use of PPAR gamma ligands during pregnancy.


Subject(s)
PPAR gamma/physiology , Placenta/metabolism , Animals , Cell Differentiation , Female , Ligands , Lipid Metabolism , Mice , PPAR gamma/genetics , Parturition/metabolism , Placenta/cytology , Placentation , Pregnancy , Transcriptional Activation , Trophoblasts/cytology , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL