Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Proc Natl Acad Sci U S A ; 117(30): 17864-17875, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32669432

ABSTRACT

Early pregnancy loss affects ∼15% of all implantation-confirmed human conceptions. However, evolutionarily conserved molecular mechanisms that regulate self-renewal of trophoblast progenitors and their association with early pregnancy loss are poorly understood. Here, we provide evidence that transcription factor TEAD4 ensures survival of postimplantation mouse and human embryos by controlling self-renewal and stemness of trophoblast progenitors within the placenta primordium. In an early postimplantation mouse embryo, TEAD4 is selectively expressed in trophoblast stem cell-like progenitor cells (TSPCs), and loss of Tead4 in postimplantation mouse TSPCs impairs their self-renewal, leading to embryonic lethality before embryonic day 9.0, a developmental stage equivalent to the first trimester of human gestation. Both TEAD4 and its cofactor, yes-associated protein 1 (YAP1), are specifically expressed in cytotrophoblast (CTB) progenitors of a first-trimester human placenta. We also show that a subset of unexplained recurrent pregnancy losses (idiopathic RPLs) is associated with impaired TEAD4 expression in CTB progenitors. Furthermore, by establishing idiopathic RPL patient-specific human trophoblast stem cells (RPL-TSCs), we show that loss of TEAD4 is associated with defective self-renewal in RPL-TSCs and rescue of TEAD4 expression restores their self-renewal ability. Unbiased genomics studies revealed that TEAD4 directly regulates expression of key cell cycle genes in both mouse and human TSCs and establishes a conserved transcriptional program. Our findings show that TEAD4, an effector of the Hippo signaling pathway, is essential for the establishment of pregnancy in a postimplantation mammalian embryo and indicate that impairment of the Hippo signaling pathway could be a molecular cause for early human pregnancy loss.


Subject(s)
Cell Self Renewal/genetics , DNA-Binding Proteins/genetics , Embryonic Development/genetics , Muscle Proteins/genetics , Transcription Factors/genetics , Trophoblasts/cytology , Trophoblasts/metabolism , Abortion, Habitual/etiology , Abortion, Habitual/metabolism , Abortion, Spontaneous/etiology , Abortion, Spontaneous/metabolism , Animals , Biomarkers , DNA-Binding Proteins/metabolism , Disease Models, Animal , Disease Susceptibility , Embryo Implantation , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Mice , Muscle Proteins/metabolism , Placenta/metabolism , Pregnancy , TEA Domain Transcription Factors , Transcription Factors/metabolism
2.
Proc Natl Acad Sci U S A ; 117(25): 14280-14291, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513715

ABSTRACT

In utero mammalian development relies on the establishment of the maternal-fetal exchange interface, which ensures transportation of nutrients and gases between the mother and the fetus. This exchange interface is established via development of multinucleated syncytiotrophoblast cells (SynTs) during placentation. In mice, SynTs develop via differentiation of the trophoblast stem cell-like progenitor cells (TSPCs) of the placenta primordium, and in humans, SynTs are developed via differentiation of villous cytotrophoblast (CTB) progenitors. Despite the critical need in pregnancy progression, conserved signaling mechanisms that ensure SynT development are poorly understood. Herein, we show that atypical protein kinase C iota (PKCλ/ι) plays an essential role in establishing the SynT differentiation program in trophoblast progenitors. Loss of PKCλ/ι in the mouse TSPCs abrogates SynT development, leading to embryonic death at approximately embryonic day 9.0 (E9.0). We also show that PKCλ/ι-mediated priming of trophoblast progenitors for SynT differentiation is a conserved event during human placentation. PKCλ/ι is selectively expressed in the first-trimester CTBs of a developing human placenta. Furthermore, loss of PKCλ/ι in CTB-derived human trophoblast stem cells (human TSCs) impairs their SynT differentiation potential both in vitro and after transplantation in immunocompromised mice. Our mechanistic analyses indicate that PKCλ/ι signaling maintains expression of GCM1, GATA2, and PPARγ, which are key transcription factors to instigate SynT differentiation programs in both mouse and human trophoblast progenitors. Our study uncovers a conserved molecular mechanism, in which PKCλ/ι signaling regulates establishment of the maternal-fetal exchange surface by promoting trophoblast progenitor-to-SynT transition during placentation.


Subject(s)
Cell Differentiation/physiology , Isoenzymes/metabolism , Maternal-Fetal Exchange/physiology , Placenta/metabolism , Protein Kinase C/metabolism , Trophoblasts/physiology , Animals , DNA-Binding Proteins/metabolism , Female , GATA2 Transcription Factor/metabolism , Humans , Isoenzymes/genetics , Male , Mice , Mice, Knockout , Models, Animal , PPAR gamma/metabolism , Placenta/cytology , Placentation/physiology , Pregnancy , Protein Kinase C/genetics , Signal Transduction , Stem Cells/cytology , Transcription Factors/metabolism , Trophoblasts/cytology
3.
Development ; 145(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30201685

ABSTRACT

Early mammalian development is crucially dependent on the establishment of oxidative energy metabolism within the trophectoderm (TE) lineage. Unlike the inner cell mass, TE cells enhance ATP production via mitochondrial oxidative phosphorylation (OXPHOS) and this metabolic preference is essential for blastocyst maturation. However, molecular mechanisms that regulate establishment of oxidative energy metabolism in TE cells are incompletely understood. Here, we show that conserved transcription factor TEAD4, which is essential for pre-implantation mammalian development, regulates this process by promoting mitochondrial transcription. In developing mouse TE and TE-derived trophoblast stem cells (TSCs), TEAD4 localizes to mitochondria, binds to mitochondrial DNA (mtDNA) and facilitates its transcription by recruiting mitochondrial RNA polymerase (POLRMT). Loss of TEAD4 impairs recruitment of POLRMT, resulting in reduced expression of mtDNA-encoded electron transport chain components, thereby inhibiting oxidative energy metabolism. Our studies identify a novel TEAD4-dependent molecular mechanism that regulates energy metabolism in the TE lineage to ensure mammalian development.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Energy Metabolism , Mammals/embryology , Mammals/genetics , Mitochondria/genetics , Muscle Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Blastocyst/cytology , Blastocyst/metabolism , Blastocyst/ultrastructure , DNA, Mitochondrial/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Directed RNA Polymerases/metabolism , Ectoderm/cytology , Electron Transport , Energy Metabolism/genetics , Mice , Mitochondria/ultrastructure , Models, Biological , Muscle Proteins/deficiency , Muscle Proteins/genetics , Oxidation-Reduction , Stem Cells/cytology , Stem Cells/metabolism , TEA Domain Transcription Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Trophoblasts/cytology
4.
J Biol Chem ; 294(46): 17301-17313, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31591264

ABSTRACT

A successful pregnancy is critically dependent upon proper placental development and function. During human placentation, villous cytotrophoblast (CTB) progenitors differentiate to form syncytiotrophoblasts (SynTBs), which provide the exchange surface between the mother and fetus and secrete hormones to ensure proper progression of pregnancy. However, epigenetic mechanisms that regulate SynTB differentiation from CTB progenitors are incompletely understood. Here, we show that lysine-specific demethylase 1 (LSD1; also known as KDM1A), a histone demethylase, is essential to this process. LSD1 is expressed both in CTB progenitors and differentiated SynTBs in first-trimester placental villi; accordingly, expression in SynTBs is maintained throughout gestation. Impairment of LSD1 function in trophoblast progenitors inhibits induction of endogenous retrovirally encoded genes SYNCYTIN1/endogenous retrovirus group W member 1, envelope (ERVW1) and SYNCYTIN2/endogenous retrovirus group FRD member 1, envelope (ERVFRD1), encoding fusogenic proteins critical to human trophoblast syncytialization. Loss of LSD1 also impairs induction of chorionic gonadotropin α (CGA) and chorionic gonadotropin ß (CGB) genes, which encode α and ß subunits of human chorionic gonadotrophin (hCG), a hormone essential to modulate maternal physiology during pregnancy. Mechanistic analyses at the endogenous ERVW1, CGA, and CGB loci revealed a regulatory axis in which LSD1 induces demethylation of repressive histone H3 lysine 9 dimethylation (H3K9Me2) and interacts with transcription factor GATA2 to promote RNA polymerase II (RNA-POL-II) recruitment and activate gene transcription. Our study reveals a novel LSD1-GATA2 axis, which regulates human trophoblast syncytialization.


Subject(s)
Cell Differentiation/genetics , GATA2 Transcription Factor/genetics , Histone Demethylases/genetics , Trophoblasts/metabolism , Chorionic Villi/growth & development , Chorionic Villi/metabolism , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation, Developmental/genetics , Gene Products, env/genetics , Humans , Mother-Child Relations , Placentation/genetics , Pregnancy , Pregnancy Proteins/genetics , RNA Polymerase II/genetics , Signal Transduction/genetics
5.
Development ; 144(5): 876-888, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28232602

ABSTRACT

GATA transcription factors are implicated in establishing cell fate during mammalian development. In early mammalian embryos, GATA3 is selectively expressed in the extraembryonic trophoblast lineage and regulates gene expression to promote trophoblast fate. However, trophoblast-specific GATA3 function is dispensable for early mammalian development. Here, using dual conditional knockout mice, we show that genetic redundancy of Gata3 with paralog Gata2 in trophoblast progenitors ensures the successful progression of both pre- and postimplantation mammalian development. Stage-specific gene deletion in trophoblasts reveals that loss of both GATA genes, but not either alone, leads to embryonic lethality prior to the onset of their expression within the embryo proper. Using ChIP-seq and RNA-seq analyses, we define the global targets of GATA2/GATA3 and show that they directly regulate a large number of common genes to orchestrate stem versus differentiated trophoblast fate. In trophoblast progenitors, GATA factors directly regulate BMP4, Nodal and Wnt signaling components that promote embryonic-extraembryonic signaling cross-talk, which is essential for the development of the embryo proper. Our study provides genetic evidence that impairment of trophoblast-specific GATA2/GATA3 function could lead to early pregnancy failure.


Subject(s)
GATA2 Transcription Factor/physiology , GATA3 Transcription Factor/physiology , Placenta/physiology , Stem Cells/cytology , Trophoblasts/cytology , Animals , Cell Differentiation , Cell Lineage , Embryo Implantation , Embryonic Development , Female , Gene Deletion , Humans , Mice , Mice, Knockout , Pregnancy , Pregnancy, Animal , Sequence Analysis, RNA
6.
Stem Cells ; 32(11): 2880-92, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142417

ABSTRACT

Pluripotent stem cells (PSCs) contain functionally immature mitochondria and rely upon high rates of glycolysis for their energy requirements. Thus, altered mitochondrial function and promotion of aerobic glycolysis are key to maintain and induce pluripotency. However, signaling mechanisms that regulate mitochondrial function and reprogram metabolic preferences in self-renewing versus differentiated PSC populations are poorly understood. Here, using murine embryonic stem cells (ESCs) as a model system, we demonstrate that atypical protein kinase C isoform, PKC lambda/iota (PKCλ/ι), is a key regulator of mitochondrial function in ESCs. Depletion of PKCλ/ι in ESCs maintains their pluripotent state as evident from germline offsprings. Interestingly, loss of PKCλ/ι in ESCs leads to impairment in mitochondrial maturation, organization, and a metabolic shift toward glycolysis under differentiating condition. Our mechanistic analyses indicate that a PKCλ/ι-hypoxia-inducible factor 1α-PGC1α axis regulates mitochondrial respiration and balances pluripotency in ESCs. We propose that PKCλ/ι could be a crucial regulator of mitochondrial function and energy metabolism in stem cells and other cellular contexts.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Energy Metabolism/physiology , Isoenzymes/metabolism , Mitochondria/metabolism , Pluripotent Stem Cells/metabolism , Protein Kinase C/metabolism , Animals , Glycolysis/physiology , Humans , Mice , Signal Transduction/physiology
7.
Proc Natl Acad Sci U S A ; 109(19): 7362-7, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22529382

ABSTRACT

In the preimplantation mouse embryo, TEAD4 is critical to establishing the trophectoderm (TE)-specific transcriptional program and segregating TE from the inner cell mass (ICM). However, TEAD4 is expressed in the TE and the ICM. Thus, differential function of TEAD4 rather than expression itself regulates specification of the first two cell lineages. We used ChIP sequencing to define genomewide TEAD4 target genes and asked how transcription of TEAD4 target genes is specifically maintained in the TE. Our analyses revealed an evolutionarily conserved mechanism, in which lack of nuclear localization of TEAD4 impairs the TE-specific transcriptional program in inner blastomeres, thereby allowing their maturation toward the ICM lineage. Restoration of TEAD4 nuclear localization maintains the TE-specific transcriptional program in the inner blastomeres and prevents segregation of the TE and ICM lineages and blastocyst formation. We propose that altered subcellular localization of TEAD4 in blastomeres dictates first mammalian cell fate specification.


Subject(s)
Cell Lineage , DNA-Binding Proteins/metabolism , Muscle Proteins/metabolism , Transcription Factors/metabolism , Animals , Blastocyst/cytology , Blastocyst/metabolism , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Blastomeres/cytology , Blastomeres/metabolism , Blotting, Western , CDX2 Transcription Factor , Cattle , Cell Nucleus/metabolism , Cells, Cultured , DNA-Binding Proteins/genetics , Embryonic Stem Cells/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Macaca mulatta , Mice , Mice, Transgenic , Muscle Proteins/genetics , RNA Interference , Rats , Reverse Transcriptase Polymerase Chain Reaction , TEA Domain Transcription Factors , Transcription Factors/genetics
8.
J Biol Chem ; 288(34): 24351-62, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23846691

ABSTRACT

Embryonic stem cell (ESC) pluripotency is orchestrated by distinct signaling pathways that are often targeted to maintain ESC self-renewal or their differentiation to other lineages. We showed earlier that inhibition of PKC signaling maintains pluripotency in mouse ESCs. Therefore, in this study, we investigated the importance of protein kinase C signaling in the context of rat ESC (rESC) pluripotency. Here we show that inhibition of PKC signaling is an efficient strategy to establish and maintain pluripotent rESCs and to facilitate reprogramming of rat embryonic fibroblasts to rat induced pluripotent stem cells. The complete developmental potential of rESCs was confirmed with viable chimeras and germ line transmission. Our molecular analyses indicated that inhibition of a PKCζ-NF-κB-microRNA-21/microRNA-29 regulatory axis contributes to the maintenance of rESC self-renewal. In addition, PKC inhibition maintains ESC-specific epigenetic modifications at the chromatin domains of pluripotency genes and, thereby, maintains their expression. Our results indicate a conserved function of PKC signaling in balancing self-renewal versus differentiation of both mouse and rat ESCs and indicate that targeting PKC signaling might be an efficient strategy to establish ESCs from other mammalian species.


Subject(s)
Embryonic Stem Cells/enzymology , Pluripotent Stem Cells/enzymology , Protein Kinase C-epsilon/metabolism , Signal Transduction/physiology , Animals , Embryonic Stem Cells/cytology , Indoles/pharmacology , Maleimides/pharmacology , MicroRNAs/metabolism , NF-kappa B/metabolism , Pluripotent Stem Cells/cytology , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Rats , Signal Transduction/drug effects
9.
Stem Cells ; 29(4): 618-28, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21308862

ABSTRACT

The intricate molecular mechanisms that regulate ESC pluripotency are incompletely understood. Prior research indicated that activation of the Janus kinase-signal transducer and activator of transcription (STAT3) pathway or inhibition of extracellular signal-regulated kinase/glycogen synthase kinase 3 (ERK/GSK3) signaling maintains mouse ESC (mESC) pluripotency. Here, we demonstrate that inhibition of protein kinase C (PKC) isoforms maintains mESC pluripotency without the activation of STAT3 or inhibition of ERK/GSK3 signaling pathways. Our analyses revealed that the atypical PKC isoform, PKCζ plays an important role in inducing lineage commitment in mESCs through a PKCζ-nuclear factor kappa-light-chain-enhancer of activated B cells signaling axis. Furthermore, inhibition of PKC isoforms permits derivation of germline-competent ESCs from mouse blastocysts and also facilitates reprogramming of mouse embryonic fibroblasts toward induced pluripotent stem cells. Our results indicate that PKC signaling is critical to balancing ESC self-renewal and lineage commitment.


Subject(s)
Cell Lineage , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Protein Kinase C/metabolism , Signal Transduction , Animals , Cell Differentiation/physiology , Cellular Reprogramming , Embryonic Stem Cells/cytology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/antagonists & inhibitors , Induced Pluripotent Stem Cells , Mice , NF-kappa B/antagonists & inhibitors , Pluripotent Stem Cells/cytology , Protein Kinase C/antagonists & inhibitors , RNA Interference , STAT3 Transcription Factor/metabolism
10.
J Biol Chem ; 285(53): 41567-77, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-21041298

ABSTRACT

Angiogenesis is critically dependent on endothelial cell-specific transcriptional mechanisms. However, the molecular processes that regulate chromatin domains and thereby dictate transcription of key endothelial genes are poorly understood. Here, we report that, in endothelial cells, angiogenic signal-mediated transcriptional induction of Vegfr1 (vascular endothelial growth factor receptor 1) is dependent on the histone chaperone, HIRA (histone cell cycle regulation-defective homolog A). Our molecular analyses revealed that, in response to angiogenic signals, HIRA is induced in endothelial cells and mediates incorporation of lysine 56 acetylated histone H3.3 (H3acK56) at the chromatin domain of Vegfr1. HIRA-mediated incorporation of H3acK56 is a general mechanism associated with transcriptional induction of several angiogenic genes in endothelial cells. Depletion of HIRA inhibits H3acK56 incorporation and transcriptional induction of Vegfr1 and other angiogenic genes. Our functional analyses revealed that depletion of HIRA abrogates endothelial network formation on Matrigel and inhibits angiogenesis in an in vivo Matrigel plug assay. Furthermore, analysis in a laser-induced choroidal neovascularization model showed that depletion of HIRA significantly inhibits neovascularization. Our results for the first time decipher a histone chaperone (HIRA)-dependent molecular mechanism in endothelial gene regulation and indicate that histone chaperones could be new targets for angiogenesis therapy.


Subject(s)
Chromatin/chemistry , Endothelium, Vascular/metabolism , Histones/chemistry , Lysine/chemistry , Animals , Collagen/chemistry , Drug Combinations , Endothelial Cells/cytology , Female , Humans , Laminin/chemistry , Mice , Mice, Inbred C57BL , Molecular Chaperones/chemistry , Neovascularization, Pathologic , Proteoglycans/chemistry , Vascular Endothelial Growth Factor Receptor-1/chemistry
11.
J Biol Chem ; 284(42): 28729-37, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19700764

ABSTRACT

During early mammalian development, genesis of the first two cell lineages, inner cell mass (ICM) and trophectoderm (TE), is dependent upon functions of key transcription factors that are expressed in a regulated and spatially restricted fashion. In this study, we demonstrate that during early mouse development, mRNA expression of transcription factor GATA3 is induced at the 4-cell stage and is consistently present during pre-implantation embryonic development. Interestingly, at the blastocyst stage, Gata3 mRNA is selectively up-regulated within the TE lineage, and GATA3 protein is abundantly present only in the TE but not in the ICM. Using mouse trophoblast stem cells (TS cells) as a model, we found that, knockdown of GATA3 by RNA interference (RNAi) down-regulates expression of caudal-type homeobox 2 (CDX2), a key regulator of the TE lineage. Chromatin immunoprecipitation (ChIP) analyses revealed that, in TS cells, GATA3 directly regulates Cdx2 transcription from a conserved GATA motif at the intron 1 region of the Cdx2 locus. ChIP analyses with mouse blastocysts also detected GATA3 occupancy at intron 1 of the Cdx2 locus. In addition, down-regulation of GATA3 in pre-implantation mouse embryos reduces Cdx2 expression and inhibits morula to blastocyst transformation. Our results indicate a novel function of GATA3, in which it is selectively expressed in TE, regulates expression of key genes in TE lineage, and is involved in morula to blastocyst transformation.


Subject(s)
Ectoderm/metabolism , Embryo Implantation/genetics , GATA3 Transcription Factor/biosynthesis , GATA3 Transcription Factor/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Trophoblasts/metabolism , Animals , Blastocyst/metabolism , CDX2 Transcription Factor , Cell Culture Techniques , Cell Lineage , Chromatin Immunoprecipitation , Humans , Mice , RNA, Messenger/metabolism , Time Factors
12.
Nucleic Acids Res ; 36(17): 5552-61, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18755708

ABSTRACT

Proteins that participate in the import of cytosolic tRNAs into mitochondria have been identified in several eukaryotic species, but the details of their interactions with tRNA and other proteins are unknown. In the kinetoplastid protozoon Leishmania tropica, multiple proteins are organized into a functional import complex. RIC8A, a tRNA-binding subunit of this complex, has a C-terminal domain that functions as subunit 6b of ubiquinol cytochrome c reductase (complex III). We show that the N-terminal domain, unique to kinetoplastid protozoa, is structurally similar to the appended S15/NS1 RNA-binding domain of aminoacyl tRNA synthetases, with a helix-turn-helix motif. Structure-guided mutagenesis coupled with in vitro assays showed that helix alpha1 contacts tRNA whereas helix alpha2 targets the protein for assembly into the import complex. Inducible expression of a helix 1-deleted variant in L. tropica resulted in formation of an inactive import complex, while the helix 2-deleted variant was unable to assemble in vivo. Moreover, a protein-interaction assay showed that the C-terminal domain makes allosteric contacts with import receptor RIC1 complexed with tRNA. These results help explain the origin of the bifunctionality of RIC8A, and the allosteric changes accompanying docking and release of tRNA during import.


Subject(s)
Leishmania tropica/metabolism , Mitochondrial Proteins/chemistry , Nucleobase, Nucleoside, Nucleotide, and Nucleic Acid Transport Proteins/chemistry , RNA, Transfer/metabolism , RNA-Binding Proteins/chemistry , Allosteric Site , Amino Acid Sequence , Animals , Leishmania tropica/genetics , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Mutagenesis , Nucleobase, Nucleoside, Nucleotide, and Nucleic Acid Transport Proteins/genetics , Nucleobase, Nucleoside, Nucleotide, and Nucleic Acid Transport Proteins/metabolism , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Transport , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Structural Homology, Protein
13.
Placenta ; 60 Suppl 1: S61-S66, 2017 12.
Article in English | MEDLINE | ID: mdl-28526138

ABSTRACT

Mammalian reproduction is critically dependent on trophoblast cells, which ensure embryo implantation and placentation. Development of trophoblast cell lineages is a multi-step process and relies upon proper spatial and temporal gene expression, which is regulated by multiple transcription factors. However, most of the transcription factors that are implicated in trophoblast development regulate gene expression at a specific developmental stage or in a specific trophoblast subtype. In contrast, recent studies from our group and other laboratories indicate that conserved GATA family of transcription factors, GATA2 and GATA3, are important to regulate gene expression at multiple stages of trophoblast development. Furthermore, our conditional gene deletion studies revealed that functional redundancy of GATA2 and GATA3 ensures both self-renewal of trophoblast stem and progenitor cells and their differentiation to trophoblast cells of a matured placenta. Together these findings indicate that GATA2/GATA3 are the master orchestrators of gene expression in trophoblast cells and they fine tune gene regulatory network to establish distinct trophoblast cell types during placentation.


Subject(s)
Embryonic Stem Cells/metabolism , GATA Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Placentation , Trophoblasts/metabolism , Animals , Blastocyst/cytology , Blastocyst/metabolism , Blastocyst/pathology , Embryonic Development , Embryonic Stem Cells/cytology , Embryonic Stem Cells/pathology , Female , Humans , Placenta/cytology , Placenta/metabolism , Placenta/pathology , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Complications/pathology , Trophoblasts/cytology , Trophoblasts/pathology
14.
Mol Cell Biol ; 33(14): 2691-705, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23671187

ABSTRACT

The first mammalian cell lineage commitment is the formation of the trophectoderm (TE) and the inner cell mass (ICM) lineages during preimplantation development. Proper development of the TE and ICM lineages is dependent upon establishment of specific transcriptional programs. However, the epigenetic mechanisms that functionally contribute to establish TE- and ICM-specific transcriptional programs are poorly understood. Here, we show that proper development of the TE and ICM lineages is coordinated via combinatorial regulation of embryonic ectoderm development (EED) and lysine-specific demethylase 6B (KDM6B). During blastocyst formation, the relative levels of EED and KDM6B expression determine altered polycomb repressor 2 (PRC2) complex recruitment and incorporation of the repressive histone H3 lysine 27 trimethylation (H3K27Me3) mark at the chromatin domains of TE-specific master regulators CDX2 and GATA3, leading to their activation in the TE lineage and repression in the ICM lineage. Furthermore, ectopic gain of EED along with depletion of KDM6B in preimplantation mouse embryos abrogates CDX2 and GATA3 expression in the nascent TE lineage. The loss of CDX2 and GATA3 in the nascent TE lineage results in improper TE development, leading to failure in embryo implantation to the uterus. Our study delineates a novel epigenetic mechanism that orchestrates proper development of the first mammalian cell lineages.


Subject(s)
Cell Lineage , Ectoderm/cytology , Jumonji Domain-Containing Histone Demethylases/metabolism , Polycomb Repressive Complex 2/metabolism , Trophoblasts/metabolism , Animals , CDX2 Transcription Factor , Chromatin/metabolism , Embryo Implantation , Embryonic Stem Cells , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Pregnancy , Protein Binding , Rats , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
15.
EMBO Rep ; 8(6): 589-95, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17510656

ABSTRACT

The mechanism of active transport of transfer RNA (tRNA) across membranes is largely unknown. Factors mediating the import of tRNA into the kinetoplast mitochondrion of the protozoon Leishmania tropica are organized into a multiprotein RNA import complex (RIC) at the inner membrane. Here, we present the complete characterization of the identities and functions of the subunits of this complex. The complex contains three mitochondrion- and eight nuclear-encoded subunits; six of the latter are necessary and sufficient for import. Antisense-mediated knockdown of essential subunits resulted in the depletion of mitochondrial tRNAs and inhibition of organellar translation. Functional complexes were reconstituted with recombinant subunits expressed in Escherichia coli. Several essential RIC subunits are identical to specific subunits of respiratory complexes. These findings provide new information on the evolution of tRNA import and the foundation for detailed structural and mechanistic studies.


Subject(s)
Leishmania tropica/metabolism , Mitochondria/genetics , Protozoan Proteins/metabolism , RNA Transport , RNA, Transfer/metabolism , Animals , Protein Subunits/metabolism , RNA, Transfer/genetics , Recombinant Proteins/metabolism
16.
Proc Natl Acad Sci U S A ; 103(22): 8354-9, 2006 05 30.
Article in English | MEDLINE | ID: mdl-16714384

ABSTRACT

In kinetoplastid protozoa, import of cytosolic tRNAs into mitochondria occurs through tRNAs interacting with membrane-bound proteins, the identities of which are unknown. The inner membrane RNA import complex of Leishmania tropica contains multiple proteins and is active for import in vitro. RIC1, the largest subunit of this complex, is structurally homologous to the conserved alpha subunit of F1 ATP synthase. The RIC1 gene complemented an atpA mutation in Escherichia coli. Antisense-mediated knockdown of RIC1/F1alpha in Leishmania resulted in depletion of several mitochondrial tRNAs belonging to distinct subsets (types I and II) that interact cooperatively or antagonistically within the import complex. The knockdown-induced defect in import of type I tRNAs was rectified in a reconstituted system by purified RIC1/F1alpha alone, but recovery of type II tRNA import additionally required a type I tRNA. RIC1/F1alpha formed stable complexes with type I, but not type II, tRNAs through the cooperation of its nucleotide binding and C-terminal domains. Thus, RIC1/F1alpha is a type I tRNA import receptor. As expected of a bifunctional protein, RIC1/F1alpha is shared by both the import complex and by respiratory complex V. Alternative use of ancient respiratory proteins may have been an important step in the evolution of tRNA import.


Subject(s)
Leishmania tropica/cytology , Leishmania tropica/metabolism , Mitochondria/metabolism , Protozoan Proteins/metabolism , RNA, Transfer/metabolism , Animals , Biological Transport , Leishmania tropica/genetics , Mitochondria/chemistry , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Structural Homology, Protein
17.
J Biol Chem ; 281(35): 25270-7, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16825195

ABSTRACT

Transport of tRNAs across the inner mitochondrial membrane of the kinetoplastid protozoon Leishmania requires interactions with specific binding proteins (receptors) in a multi-subunit complex. The allosteric model of import regulation proposes cooperative and antagonistic interactions between two or more receptors with binding specificities for distinct tRNA families (types I and II, respectively). To identify the type II receptor, the gene encoding RIC8A, a subunit of the complex, was cloned. The C-terminal region of RIC8A is homologous to subunit 6b of ubiquinol cytochrome c reductase (respiratory complex III), while the N-terminal region has intrinsic affinity for type II, but not for type I, tRNAs. RIC8A is shared by the import complex and complex III, indicating its bi-functionality, but is assembled differently in the two complexes. Knockdown of RIC8A in Leishmania lowered the mitochondrial content of type II tRNAs but raised that of type I tRNAs, with downstream effects on mitochondrial translation and respiration, and cell death. In RIC8A knockdown cells, a subcomplex was formed that interacted with type I tRNA, but the negative regulation by type II tRNA was lost. Mitochondrial extracts from these cells were defective for type II, but not type I, import; import and regulation were restored by purified RIC8A. These results provide evidence for the relevance of allosteric regulation in vivo and indicate that acquisition of new tRNA-binding domains by ancient respiratory components have played a key role in the evolution of mitochondrial tRNA import.


Subject(s)
Guanine Nucleotide Exchange Factors/chemistry , Leishmania tropica/metabolism , Membrane Transport Proteins/chemistry , Mitochondria/metabolism , RNA, Transfer/chemistry , RNA/metabolism , Allosteric Site , Animals , Biological Transport , Cloning, Molecular , Cross-Linking Reagents/pharmacology , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/physiology , Humans , Membrane Transport Proteins/physiology , Models, Molecular , Protein Structure, Tertiary , Recombinant Proteins/chemistry
18.
Differentiation ; 71(9-10): 549-56, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14686952

ABSTRACT

Differentiation of kinetoplastid protozoa during their complex life cycles is accompanied by stepwise changes in mitochondrial functions. Recent studies have begun to reveal multilevel post-transcriptional regulatory mechanisms by which the expression of the nuclear and mitochondrially encoded components of respiratory enzymes is coordinated, as well as the identities of some general and gene-specific factors controlling mitochondrial differentiation.


Subject(s)
Cell Differentiation/physiology , Kinetoplastida/cytology , Kinetoplastida/physiology , Mitochondria/genetics , RNA, Protozoan/physiology , Animals , Eukaryota/genetics , Gene Expression Regulation , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Trypanosoma/cytology , Trypanosoma/physiology
SELECTION OF CITATIONS
SEARCH DETAIL