Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(2): 113710, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38306272

ABSTRACT

Without new transcription, gene expression across the oocyte-to-embryo transition (OET) relies instead on regulation of mRNA poly(A) tails to control translation. However, how tail dynamics shape translation across the OET in mammals remains unclear. We perform long-read RNA sequencing to uncover poly(A) tail lengths across the mouse OET and, incorporating published ribosome profiling data, provide an integrated, transcriptome-wide analysis of poly(A) tails and translation across the entire transition. We uncover an extended wave of global deadenylation during fertilization in which short-tailed, oocyte-deposited mRNAs are translationally activated without polyadenylation through resistance to deadenylation. Subsequently, in the embryo, mRNAs are readenylated and translated in a surge of global polyadenylation. We further identify regulation of poly(A) tail length at the isoform level and stage-specific enrichment of mRNA sequence motifs among regulated transcripts. These data provide insight into the stage-specific mechanisms of poly(A) tail regulation that orchestrate gene expression from oocyte to embryo in mammals.


Subject(s)
Embryo, Mammalian , Gene Expression Profiling , Animals , Mice , Oocytes , RNA, Messenger/genetics , Mammals
2.
Cell Rep ; 43(2): 113701, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38277271

ABSTRACT

Human embryo implantation is remarkably inefficient, and implantation failure remains among the greatest obstacles in treating infertility. Gene expression data from human embryos have accumulated rapidly in recent years; however, identification of the subset of genes that determine successful implantation remains a challenge. We leverage clinical morphologic grading-known for decades to correlate with implantation potential-and transcriptome analyses of matched embryonic and abembryonic samples to identify factors and pathways enriched and depleted in human blastocysts of good and poor morphology. Unexpectedly, we discovered that the greatest difference was in the state of extraembryonic primitive endoderm (PrE) development, with relative deficiencies in poor morphology blastocysts. Our results suggest that implantation success is most strongly influenced by the embryonic compartment and that deficient PrE development is common among embryos with decreased implantation potential. Our study provides a valuable resource for those investigating the markers and mechanisms of human embryo implantation.


Subject(s)
Embryonic Development , Endoderm , Humans , Embryonic Development/genetics , Embryo Implantation/genetics , Blastocyst/metabolism , Embryo, Mammalian
3.
Development ; 149(21)2022 11 01.
Article in English | MEDLINE | ID: mdl-36255229

ABSTRACT

Nonsense-mediated RNA decay (NMD) is a highly conserved RNA turnover pathway that degrades RNAs harboring in-frame stop codons in specific contexts. Loss of NMD factors leads to embryonic lethality in organisms spanning the phylogenetic scale, but the mechanism remains unknown. Here, we report that the core NMD factor, UPF2, is required for expansion of epiblast cells within the inner cell mass of mice in vivo. We identify NMD target mRNAs in mouse blastocysts - both canonical and alternatively processed mRNAs - including those encoding cell cycle arrest and apoptosis factors, raising the possibility that NMD is essential for embryonic cell proliferation and survival. In support, the inner cell mass of Upf2-null blastocysts rapidly regresses with outgrowth and is incompetent for embryonic stem cell derivation in vitro. In addition, we uncovered concordant temporal- and lineage-specific regulation of NMD factors and mRNA targets, indicative of a shift in NMD magnitude during peri-implantation development. Together, our results reveal developmental and molecular functions of the NMD pathway in the early embryo.


Subject(s)
Nonsense Mediated mRNA Decay , RNA , Mice , Animals , RNA/metabolism , Phylogeny , Nonsense Mediated mRNA Decay/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Germ Layers/metabolism , RNA-Binding Proteins/metabolism
4.
Dev Cell ; 44(3): 392-402.e7, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29408237

ABSTRACT

Global transcriptional silencing is a highly conserved mechanism central to the oocyte-to-embryo transition. We report the unexpected discovery that global transcriptional silencing in oocytes depends on an mRNA decay activator. Oocyte-specific loss of ZFP36L2 an RNA-binding protein that promotes AU-rich element-dependent mRNA decay prevents global transcriptional silencing and causes oocyte maturation and fertilization defects, as well as complete female infertility in the mouse. Single-cell RNA sequencing revealed that ZFP36L2 downregulates mRNAs encoding transcription and chromatin modification regulators, including a large group of mRNAs for histone demethylases targeting H3K4 and H3K9, which we show are bound and degraded by ZFP36L2. Oocytes lacking Zfp36l2 fail to accumulate histone methylation at H3K4 and H3K9, marks associated with the transcriptionally silent, developmentally competent oocyte state. Our results uncover a ZFP36L2-dependent mRNA decay mechanism that acts as a developmental switch during oocyte growth, triggering wide-spread shifts in chromatin modification and global transcription.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation, Developmental , Gene Silencing , Infertility, Female/pathology , Oocytes/metabolism , Transcription, Genetic , Tristetraprolin/physiology , Animals , Female , High-Throughput Nucleotide Sequencing , Infertility, Female/genetics , Infertility, Female/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oogenesis/genetics , RNA Stability/genetics , Single-Cell Analysis , Transcriptome
5.
Cell Cycle ; 12(10): 1521-35, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23598720

ABSTRACT

TGF-ß1 plays biphasic functions in prostate tumorigenesis, inhibiting cell growth at early stages but promoting malignant progression at later stages. However, the molecular basis for the oncogenic conversion of TGF-ß1 function remains largely undefined. Here, we demonstrate that PPARδ is a direct transcription target of TGF-ß1 and plays a critical role in oncogenic redirection of TGF-ß1 signaling. Blockade of PPARδ induction enhances tumor cell response to TGF-ß1-mediated growth inhibition, while its activation promotes TGF-ß1-induced tumor growth, migration and invasion. PPARδ-mediated switch of TGF-ß1 function is associated with down- and upregulation of Smad and ERK signaling, respectively, and tightly linked to its function to activate ABCA1 cholesterol transporter followed by caveolin-1 (Cav1) induction. Intriguingly, TGF-ß1 activation of the PPARδ-ABCA1-Cav1 pathway facilitates degradation of TGF-ß receptors (TßRs) and attenuates Smad but enhances ERK response to TGF-ß1. Expression of PPARδ and Cav1 is tightly correlated in both prostate tissues and cell lines and significantly higher in cancer vs. normal tissues. Collectively, our study shows that PPARδ is a transcription target of TGF-ß1 and contributes to the oncogenic conversion of TGF-ß1 function through activation of the ABCA1-Cav1-TßR signaling axis.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Caveolin 1/metabolism , PPAR delta/metabolism , Transforming Growth Factor beta1/metabolism , ATP Binding Cassette Transporter 1/antagonists & inhibitors , ATP Binding Cassette Transporter 1/genetics , Animals , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Cell Line, Tumor , Cell Movement , Humans , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , PPAR delta/antagonists & inhibitors , PPAR delta/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Smad Proteins/antagonists & inhibitors , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/pharmacology , Transplantation, Heterologous
6.
PLoS One ; 7(4): e34342, 2012.
Article in English | MEDLINE | ID: mdl-22496792

ABSTRACT

The p120-catenin family has undergone a significant expansion during the evolution of vertebrates, resulting in varied functions that have yet to be discerned or fully characterized. Likewise, members of the plakophilins, a related catenin subfamily, are found throughout the cell with little known about their functions outside the desmosomal plaque. While the plakophilin-3 (Pkp3) knockout mouse resulted in skin defects, we find larger, including lethal effects following its depletion in Xenopus. Pkp3, unlike some other characterized catenins in amphibians, does not have significant maternal deposits of mRNA. However, during embryogenesis, two Pkp3 protein products whose temporal expression is partially complimentary become expressed. Only the smaller of these products is found in adult Xenopus tissues, with an expression pattern exhibiting distinctions as well as overlaps with those observed in mammalian studies. We determined that Xenopus Pkp3 depletion causes a skin fragility phenotype in keeping with the mouse knockout, but more novel, Xenopus tailbud embryos are hyposensitive to touch even in embryos lacking outward discernable phenotypes, and we additionally resolved disruptions in certain peripheral neural structures, altered establishment and migration of neural crest, and defects in ectodermal multiciliated cells. The use of two distinct morpholinos, as well as rescue approaches, indicated the specificity of these effects. Our results point to the requirement of Pkp3 in amphibian embryogenesis, with functional roles in a number of tissue types.


Subject(s)
Ectoderm/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Neural Crest/metabolism , Plakophilins/physiology , Xenopus laevis/embryology , Amino Acid Sequence , Animals , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Catenins/genetics , Catenins/metabolism , Cell Movement , Desmosomes/metabolism , Ectoderm/cytology , Female , In Situ Hybridization , Mice , Molecular Sequence Data , Neural Crest/cytology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Sequence Homology, Amino Acid , Xenopus laevis/metabolism
7.
PLoS One ; 7(1): e30814, 2012.
Article in English | MEDLINE | ID: mdl-22303459

ABSTRACT

Wnt signaling is crucial for a variety of biological processes, including body axis formation, planar polarity, stem cell maintenance and cellular differentiation. Therefore, targeted manipulation of Wnt signaling in vivo would be extremely useful. By applying chemical inducer of dimerization (CID) technology, we were able to modify the Wnt co-receptor, low-density lipoprotein (LDL)-receptor-related protein 5 (LRP5), to generate the synthetic ligand inducible Wnt switch, iLRP5. We show that iLRP5 oligomerization results in its localization to disheveled-containing punctate structures and sequestration of scaffold protein Axin, leading to robust ß-catenin-mediated signaling. Moreover, we identify a novel LRP5 cytoplasmic domain critical for its intracellular localization and casein kinase 1-dependent ß-catenin signaling. Finally, by utilizing iLRP5 as a Wnt signaling switch, we generated the Ubiquitous Activator of ß-catenin (Ubi-Cat) transgenic mouse line. The Ubi-Cat line allows for nearly ubiquitous expression of iLRP5 under control of the H-2K(b) promoter. Activation of iLRP5 in isolated prostate basal epithelial stem cells resulted in expansion of p63(+) cells and development of hyperplasia in reconstituted murine prostate grafts. Independently, iLRP5 induction in adult prostate stroma enhanced prostate tissue regeneration. Moreover, induction of iLRP5 in male Ubi-Cat mice resulted in prostate tumor progression over several months from prostate hyperplasia to adenocarcinoma. We also investigated iLRP5 activation in Ubi-Cat-derived mammary cells, observing that prolonged activation results in mammary tumor formation. Thus, in two distinct experimental mouse models, activation of iLRP5 results in disruption of tissue homeostasis, demonstrating the utility of iLRP5 as a novel research tool for determining the outcome of Wnt activation in a precise spatially and temporally determined fashion.


Subject(s)
Homeostasis , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Protein Multimerization , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Axin Protein/metabolism , Casein Kinase I/metabolism , Dishevelled Proteins , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Intracellular Space/metabolism , Male , Mammary Neoplasms, Animal/pathology , Membrane Microdomains/metabolism , Mice , Molecular Sequence Data , Phosphoproteins/metabolism , Phosphorylation , Prostate/pathology , Prostate/transplantation , Protein Binding , Protein Transport , Stromal Cells/metabolism , Stromal Cells/pathology , Structure-Activity Relationship , Tumor Suppressor Proteins/metabolism , beta Catenin/metabolism
8.
Dev Dyn ; 240(12): 2601-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22028074

ABSTRACT

The novel adaptor protein Kazrin associates with multifunctional entities including p120-subfamily members (ARVCF-, delta-, and p0071-catenin). Critical contributions of Kazrin to development or homeostasis are indicated with respect to ectoderm formation, integrity and keratinocyte differentiation, whereas its presence in varied tissues suggests broader roles. We find that Kazrin is maternally loaded, is expressed across development and becomes enriched in the forming head. Kazrin's potential contributions to craniofacial development were probed by means of knockdown in the prospective anterior neural region. Cartilaginous head structures as well as eyes on injected sides were reduced in size, with molecular markers suggesting an impact upon neural crest cell establishment and migration. Similar effects followed the depletion of ARVCF (or delta-catenin), with Kazrin:ARVCF functional interplay supported upon ARVCF partial rescue of Kazrin knockdown phenotypes. Thus, Kazrin and its associating ARVCF- and delta-catenins, are required to form craniofacial tissues originating from cranial neural crest and precordal plate.


Subject(s)
Armadillo Domain Proteins/metabolism , Catenins/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Membrane Proteins/metabolism , Neural Crest/embryology , Organogenesis/physiology , Phosphoproteins/metabolism , Skull/embryology , Xenopus Proteins/metabolism , Animals , Armadillo Domain Proteins/genetics , Cartilage/embryology , Catenins/genetics , Cell Adhesion Molecules/genetics , Eye/embryology , Gene Knockdown Techniques , Membrane Proteins/genetics , Phosphoproteins/genetics , Skull/metabolism , Xenopus Proteins/genetics , Xenopus laevis , Delta Catenin
9.
J Am Soc Nephrol ; 22(9): 1654-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21804089

ABSTRACT

Canonical ß-catenin-mediated Wnt signaling is essential for the induction of nephron development. Noncanonical Wnt/planar cell polarity (PCP) pathways contribute to processes such as cell polarization and cytoskeletal modulation in several tissues. Although PCP components likely establish the plane of polarization in kidney tubulogenesis, whether PCP effectors directly modulate the actin cytoskeleton in tubulogenesis is unknown. Here, we investigated the roles of Wnt PCP components in cytoskeletal assembly during kidney tubule morphogenesis in Xenopus laevis and zebrafish. We found that during tubulogenesis, the developing pronephric anlagen expresses Daam1 and its interacting Rho-GEF (WGEF), which compose one PCP/noncanonical Wnt pathway branch. Knockdown of Daam1 resulted in reduced expression of late pronephric epithelial markers with no apparent effect upon early markers of patterning and determination. Inhibiting various points in the Daam1 signaling pathway significantly reduced pronephric tubulogenesis. These data indicate that pronephric tubulogenesis requires the Daam1/WGEF/Rho PCP pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Polarity , Cytoskeleton/metabolism , Kidney Tubules/embryology , Organogenesis , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Apoptosis , Cell Proliferation , Female , Guanine Nucleotide Exchange Factors/metabolism , Xenopus laevis , Zebrafish , Zebrafish Proteins/metabolism
10.
J Cell Sci ; 123(Pt 24): 4351-65, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21098636

ABSTRACT

Wnt signaling pathways have fundamental roles in animal development and tumor progression. Here, employing Xenopus embryos and mammalian cell lines, we report that the degradation machinery of the canonical Wnt pathway modulates p120-catenin protein stability through mechanisms shared with those regulating ß-catenin. For example, in common with ß-catenin, exogenous expression of destruction complex components, such as GSK3ß and axin, promotes degradation of p120-catenin. Again in parallel with ß-catenin, reduction of canonical Wnt signals upon depletion of LRP5 and LRP6 results in p120-catenin degradation. At the primary sequence level, we resolved conserved GSK3ß phosphorylation sites in the amino-terminal region of p120-catenin present exclusively in isoform-1. Point-mutagenesis of these residues inhibited the association of destruction complex components, such as those involved in ubiquitylation, resulting in stabilization of p120-catenin. Functionally, in line with predictions, p120 stabilization increased its signaling activity in the context of the p120-Kaiso pathway. Importantly, we found that two additional p120-catenin family members, ARVCF-catenin and δ-catenin, associate with axin and are degraded in its presence. Thus, as supported using gain- and loss-of-function approaches in embryo and cell line systems, canonical Wnt signals appear poised to have an impact upon a breadth of catenin biology in vertebrate development and, possibly, human cancers.


Subject(s)
Catenins/metabolism , Signal Transduction , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Axin Protein , Casein Kinase I/metabolism , Catenins/chemistry , Cell Line , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Molecular Sequence Data , Mutant Proteins/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Interaction Mapping , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Protein Stability , Protein Structure, Tertiary , Repressor Proteins/metabolism , Ubiquitin/metabolism , Ubiquitination , Xenopus , Xenopus Proteins , Delta Catenin
11.
J Cell Sci ; 123(Pt 23): 4128-44, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21062899

ABSTRACT

In common with other p120-catenin subfamily members, Xenopus ARVCF (xARVCF) binds cadherin cytoplasmic domains to enhance cadherin metabolic stability or, when dissociated, modulates Rho-family GTPases. We report here that xARVCF binds and is stabilized by Xenopus KazrinA (xKazrinA), a widely expressed conserved protein that bears little homology to established protein families, and which is known to influence keratinocyte proliferation and differentiation and cytoskeletal activity. Although we found that xKazrinA binds directly to xARVCF, we did not resolve xKazrinA within a larger ternary complex with cadherin, nor did it co-precipitate with core desmosomal components. Instead, screening revealed that xKazrinA binds spectrin, suggesting a potential means by which xKazrinA localizes to cell-cell borders. This was supported by the resolution of a ternary biochemical complex of xARVCF-xKazrinA-xß2-spectrin and, in vivo, by the finding that ectodermal shedding followed depletion of xKazrin in Xenopus embryos, a phenotype partially rescued with exogenous xARVCF. Cell shedding appeared to be the consequence of RhoA activation, and thereby altered actin organization and cadherin function. Indeed, we also revealed that xKazrinA binds p190B RhoGAP, which was likewise capable of rescuing Kazrin depletion. Finally, xKazrinA was found to associate with δ-catenins and p0071-catenins but not with p120-catenin, suggesting that Kazrin interacts selectively with additional members of the p120-catenin subfamily. Taken together, our study supports the essential role of Kazrin in development, and reveals the biochemical and functional association of KazrinA with ARVCF-catenin, spectrin and p190B RhoGAP.


Subject(s)
Armadillo Domain Proteins/metabolism , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , GTPase-Activating Proteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Spectrin/metabolism , Xenopus Proteins/metabolism , Xenopus/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/genetics , Cadherins/genetics , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Line , Epithelial Cells/chemistry , Epithelial Cells/enzymology , GTPase-Activating Proteins/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Binding , Protein Structure, Tertiary , Spectrin/genetics , Two-Hybrid System Techniques , Xenopus/genetics , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , rhoA GTP-Binding Protein/genetics
12.
J Cell Sci ; 122(Pt 22): 4049-61, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19843587

ABSTRACT

Catenins of the p120 subclass display an array of intracellular localizations and functions. Although the genetic knockout of mouse delta-catenin results in mild cognitive dysfunction, we found severe effects of its depletion in Xenopus. delta-catenin in Xenopus is transcribed as a full-length mRNA, or as three (or more) alternatively spliced isoforms designated A, B and C. Further structural and functional complexity is suggested by three predicted and alternative translation initiation sites. Transcript analysis suggests that each splice isoform is expressed during embryogenesis, with the B and C transcript levels varying according to developmental stage. Unlike the primarily neural expression of delta-catenin reported in mammals, delta-catenin is detectable in most adult Xenopus tissues, although it is enriched in neural structures. delta-catenin associates with classical cadherins, with crude embryo fractionations further revealing non-plasma-membrane pools that might be involved in cytoplasmic and/or nuclear functions. Depletion of delta-catenin caused gastrulation defects, phenotypes that were further enhanced by co-depletion of the related p120-catenin. Depletion was significantly rescued by titrated p120-catenin expression, suggesting that these catenins have shared roles. Biochemical assays indicated that delta-catenin depletion results in reduced cadherin levels and cell adhesion, as well as perturbation of RhoA and Rac1. Titrated doses of C-cadherin, dominant-negative RhoA or constitutively active Rac1 significantly rescued delta-catenin depletion. Collectively, our experiments indicate that delta-catenin has an essential role in amphibian development, and has functional links to cadherins and Rho-family GTPases.


Subject(s)
Cadherins/metabolism , Catenins/genetics , Catenins/metabolism , Gastrula/metabolism , Xenopus laevis/embryology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Alternative Splicing , Animals , Base Sequence , Cell Adhesion , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gastrula/ultrastructure , Gastrulation/physiology , Gene Knockout Techniques , Molecular Sequence Data , Neurulation/physiology , Xenopus laevis/metabolism , Delta Catenin
13.
Proc Natl Acad Sci U S A ; 105(19): 6936-41, 2008 May 13.
Article in English | MEDLINE | ID: mdl-18458345

ABSTRACT

The recent identification of the genes responsible for several human genetic diseases affecting bone homeostasis and the characterization of mouse models for these diseases indicated that canonical Wnt signaling plays a critical role in the control of bone mass. Here, we report that the osteoblast-specific transcription factor Osterix (Osx), which is required for osteoblast differentiation, inhibits Wnt pathway activity. First, in calvarial cells of embryonic day (E)18.5 Osx-null embryos, expression of the Wnt antagonist Dkk1 was abolished, and that of Wnt target genes c-Myc and cyclin D1 was increased. Moreover, our studies demonstrated that Osx bound to and activated the Dkk1 promoter. In addition, Osx inhibited beta-catenin-induced Topflash reporter activity and beta-catenin-induced secondary axis formation in Xenopus embryos. Importantly, in calvaria of E18.5 Osx-null embryos harboring the TOPGAL reporter transgene, beta-galactosidase activity was increased, suggesting that Osx inhibited the Wnt pathway in osteoblasts in vivo. Our data further showed that Osx disrupted binding of Tcf to DNA, providing a likely mechanism for the inhibition by Osx of beta-catenin transcriptional activity. We also showed that Osx decreased osteoblast proliferation. Indeed, E18.5 Osx-null calvaria showed greater BrdU incorporation than wild-type calvaria and that Osx overexpression in C2C12 mesenchymal cells inhibited cell growth. Because Wnt signaling has a major role in stimulating osteoblast proliferation, we speculate that Osx-mediated inhibition of osteoblast proliferation is a consequence of the Osx-mediated control of Wnt/beta-catenin activity. Our results add a layer of control to Wnt/beta-catenin signaling in bone.


Subject(s)
Osteoblasts/metabolism , Signal Transduction , Transcription Factors/metabolism , Wnt Proteins/metabolism , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Down-Regulation , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Models, Biological , Organ Specificity , Osteoblasts/cytology , Peptides/metabolism , Proline-Rich Protein Domains , Promoter Regions, Genetic/genetics , Protein Binding , Sp7 Transcription Factor , Transcription Factors/chemistry , Transcription, Genetic , Transfection , Wnt Proteins/genetics , Xenopus , beta Catenin/genetics
14.
Dev Cell ; 8(6): 843-54, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15935774

ABSTRACT

Beta-catenin-dependent or canonical Wnt signals are fundamental in animal development and tumor progression. Using Xenopus laevis, we report that the BTB/POZ zinc finger family member Kaiso directly represses canonical Wnt gene targets (Siamois, c-Fos, Cyclin-D1, and c-Myc) in conjunction with TCF/LEF (TCF). Analogous to beta-catenin relief of TCF repressive activity, we show that p120-catenin relieves Kaiso-mediated repression of Siamois. Furthermore, Kaiso and TCF coassociate, and combined Kaiso and TCF derepression results in pronounced Siamois expression and increased beta-catenin coprecipitation with the Siamois promoter. The functional interdependency is underlined by Kaiso suppression of beta-catenin-induced axis duplication and by TCF-3 rescue of Kaiso depletion phenotypes. These studies point to convergence of parallel p120-catenin/Kaiso and beta-catenin/TCF signaling pathways to regulate gene expression in vertebrate development and possibly carcinogenesis.


Subject(s)
Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic/physiology , Xenopus Proteins/metabolism , Xenopus Proteins/physiology , Animals , Blotting, Western/methods , Body Patterning/genetics , Body Patterning/physiology , Chromatin Immunoprecipitation/methods , Cloning, Molecular/methods , Dose-Response Relationship, Drug , Drug Interactions , Electrophoretic Mobility Shift Assay/methods , Fluorescent Antibody Technique/methods , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoprecipitation/methods , In Situ Hybridization/methods , Microinjections/methods , Mutation/physiology , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Xenopus Proteins/genetics , Xenopus laevis , beta Catenin
15.
Mol Cell Biol ; 25(6): 2147-57, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15743813

ABSTRACT

We performed chromatin immunoprecipitation (ChIP) analyses of developmentally staged solid tissues isolated from wild-type and p53-null mice to determine specific histone N-terminal modifications, histone-modifying proteins, and transcription factor interactions at the developmental repressor region (-850) and core promoter of the hepatic tumor marker alpha-fetoprotein (AFP) gene. Both repression of AFP during liver development and silencing in the brain, where AFP is never expressed, are associated with dimethylation of histone H3 lysine 9 (DiMetH3K9) and the presence of heterochromatin protein 1 (HP1). These heterochromatic markers remain localized to AFP during developmental repression but spread to the upstream albumin gene during silencing. Developmentally regulated decreases in levels of acetylated H3 (AcH3K9) and H4 (AcH4) and of di- and trimethylated H3K4 (DiMetH3K4 and TriMetH3K4) occur at both the core promoter and distal repressor regions of AFP. Hepatic expression of AFP correlates with FoxA interaction at the repressor region and the binding of RNA polymerase II and TATA-binding protein to the core promoter. p53 acts as a developmental repressor of AFP in the liver by binding to chromatin, excluding FoxA interaction and targeting mSin3A/HDAC1 to the distal repressor region. p53-null mice exhibit developmentally delayed AFP repression, concomitant with acetylation of H3K9, methylation of H3K4, and loss of DiMetH3K9, mSin3A/HDAC1, and HP1 interactions.


Subject(s)
Gene Expression Regulation, Developmental , Heterochromatin/metabolism , Liver/growth & development , Repressor Proteins/physiology , Tumor Suppressor Protein p53/physiology , alpha-Fetoproteins/genetics , Acetylation , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/physiology , DNA-Binding Proteins/metabolism , Gene Silencing , Hepatocyte Nuclear Factor 3-alpha , Hepatocytes/metabolism , Histones/metabolism , Liver/cytology , Liver/metabolism , Methylation , Mice , Mice, Transgenic , Mutation/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Cancer Res ; 63(21): 7068-75, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14612497

ABSTRACT

X-linked inhibitor of apoptosis (XIAP) is the most potent member of the IAP family that exerts antiapoptotic effects by interfering with the activities of caspases. Recently, XIAP-associated factor 1 (XAF1) and two mitochondrial proteins, Smac/DIABLO and HtrA2, have been identified to negatively regulate the caspase-inhibiting activity of XIAP. To explore the candidacy of XAF1, Smac/DIABLO, and HtrA2 as a tumor suppressor in gastric tumorigenesis, we investigated the expression and mutation status of the genes in 123 gastric tissues and 15 cancer cell lines. Whereas Smac/DIABLO and HtrA2 transcripts were normally expressed in all cancer specimens we examined, XAF1 transcript was not expressed or present at extremely low levels in 40% (6 of 15) of cancer cell lines and in 23% (20 of 87) of primary carcinomas. Abnormal reduction of XAF1 expression showed a strong correlation with stage and grade of tumors, and a tumor-specific down-regulation of XAF1 was observed in 45% (9 of 20) of matched sets. Unlike XAF1, XIAP expression exhibited no detectable alteration in cancers. Whereas loss of heterozygosity within the XAF1 region or somatic mutations of the gene was not detected, expression of XAF1 transcript was reactivated in all nonexpressor cell lines after 5-aza-2-deoxycytidine treatment. The 5' upstream region of the XAF1 gene encompasses no gastric cell-rich region that rigorously satisfies the formal criteria for CpG islands. However, bisulfite DNA sequencing analysis for 34 CpG sites in the promoter region revealed a strong association between hypermethylation and gene silencing. Moreover, transcriptional silencing of XAF1 was tightly associated with hypermethylation of seven CpGs located in the 5' proximal region (nucleotides -23 to -234). Additionally, loss or abnormal reduction of XAF1 expression was found to inversely correlate with p53 mutations, suggesting that epigenetic inactivation of XAF1 and mutational alteration of p53 might be mutually exclusive events in gastric tumorigenesis. Collectively, our study suggests that epigenetic silencing of XAF1 by aberrant promoter methylation may contribute to the malignant progression of human gastric tumors.


Subject(s)
Adenocarcinoma/genetics , Chromosomes, Human, Pair 17/genetics , DNA Methylation , Genes, Tumor Suppressor , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Adenocarcinoma/metabolism , Alleles , Apoptosis Regulatory Proteins , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line, Tumor , Down-Regulation , Gene Deletion , Gene Expression Regulation, Neoplastic , Gene Silencing , High-Temperature Requirement A Serine Peptidase 2 , Humans , Intracellular Signaling Peptides and Proteins , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Mutation , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Stomach Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics
17.
Oncogene ; 22(28): 4314-32, 2003 Jul 10.
Article in English | MEDLINE | ID: mdl-12853969

ABSTRACT

Transforming growth factor (TGF)-beta1 acts as a potent growth inhibitor of prostate epithelial cells, and aberrant function of its receptor type I and II correlates with tumor aggressiveness. However, intracellular and serum TGF-beta1 levels are elevated in prostate cancer patients and further increased in patients with metastatic carcinoma, suggesting the oncogenic switch of TGF-beta1 role in prostate tumorigenesis. Recently, we reported the mitogenic conversion of TGF-beta1 effect by oncogenic Ha-Ras in prostate cancer cells. Here, we show that TGF-beta1 activates interleukin (IL)-6, which has been implicated in the malignant progression of prostate cancers, via multiple signaling pathways including Smad2, nuclear factor-kappaB (NF-kappaB), JNK, and Ras. TGF-beta1-induced IL-6 gene expression was strongly inhibited by DN-Smad2 but not by DN-Smad3 while it was further activated by wild-type Smad2 transfection. IL-6 activation by TGF-beta1 was accompanied by nuclear translocation of NF-kappaB, which was blocked by the p38 inhibitors SB202190 and SB203580 or by IkappaBalphaDeltaN transfection, indicating the crucial role for the p38-NF-kappaB signaling in TGF-beta1 induction of IL-6. TGF-beta1 activated c-Jun phosphorylation, and IL-6 induction by TGF-beta1 was severely impeded by DN-c-Jun and DN-JNK or AP-1 inhibitor curcumin, showing that the JNK-c-Jun-AP-1 signaling plays a pivotal role in TGF-beta1 stimulation of IL-6. It was also found that the Ras-Raf-MEK1 cascade is activated by TGF-beta1 and participates in the TGF-beta1 induction of IL-6 in an AP-1-dependent manner. Cotransfection assays demonstrated that TGF-beta1 stimulation of IL-6 results from the synergistic collaboration of the Smad2, p38-NF-kappaB, JNK-c-Jun-AP-1, or Ras-Raf-MEK1 cascades. In addition, a time course IL-6 decay revealed that mRNA stability of IL-6 is modestly increased by TGF-beta1, indicating that TGF-beta1 also regulates IL-6 at the post-transcriptional level. Intriguingly, IL-6 inactivation restored the sensitivity to TGF-beta1-mediated growth arrest and apoptosis, suggesting that elevated IL-6 in advanced prostate tumors might act as a resistance factor against TGF-beta1. Collectively, our data demonstrate that IL-6 expression is stimulated by tumor-producing TGF-beta1 in human prostate cancer cells through multiple signaling pathways including Smad2, p38, JNK, and Ras, and enhanced expression of IL-6 could contribute to the oncogenic switch of TGF-beta1 role for prostate tumorigenesis, in part by counteracting its growth suppression function.


Subject(s)
DNA-Binding Proteins/physiology , Interleukin-6/genetics , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Prostatic Neoplasms/etiology , Trans-Activators/physiology , Transforming Growth Factor beta/pharmacology , Apoptosis , Humans , JNK Mitogen-Activated Protein Kinases , Male , RNA, Messenger/analysis , Smad2 Protein , Transcription Factor AP-1/physiology , Transforming Growth Factor beta1 , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
18.
Int J Cancer ; 104(3): 318-27, 2003 Apr 10.
Article in English | MEDLINE | ID: mdl-12569555

ABSTRACT

Mutational alterations of PTEN and PIK3CA, which negatively and positively regulate PI3-kinase activity, respectively, have been observed in many types of human cancer. To explore the implication of PTEN and PIK3CA mutations in gastric tumorigenesis, we characterized the expression and mutation status of the genes in 126 gastric tissues and 15 cell lines. Expression of PTEN transcript was abnormally low in 5 of 15 (33%) cell lines and 20 of 55 (36%) primary carcinomas, whereas 0 of 71 noncancerous tissues including 16 benign tumors showed altered expression. Allelotyping analysis using an intragenic polymorphism (IVS4+109) revealed that 14 of 30 (47%) informative cases carried LOH of the gene, which is closely linked to low expression. The LOH rate was significantly higher in advanced tumors [12 of 19 (63%)] compared to early-stage tumors [2 of 11 (18%)] and more frequent in poorly differentiated tumors [9 of 13 (69%)] than well- or moderately differentiated tumors [5 of 17 (29%)]. Interestingly, however, none of the LOH tumors carried mutational disruption of the remaining allele, suggesting haploinsufficiency of PTEN in gastric tumorigenesis. Methylation studies revealed that PTEN pseudogene, but not PTEN, is methylated in cell lines and primary tumors, indicating that PTEN is not a target of epigenetic silencing in gastric cancers and that the pseudogene should be considered more carefully in methylation analysis of the PTEN promoter. Genomic amplification of PIK3CA was found in 9 of 15 (60%) cell lines and 20 of 55 (36.4%) primary tumors but in no noncancerous tissues. Furthermore, PIK3CA amplification was predominantly detected in tumors with no PTEN alterations, suggesting that mutations of PTEN and PIK3CA are mutually exclusive events in gastric tumorigenesis. Amplification of PIK3CA was strongly associated with increased expression of PIK3CA transcript and elevated levels of phospho-AKT. Collectively, our data reveal that 13 of 15 (87%) gastric cell lines and 31 of 55 (56%) primary carcinomas harbored either amplification of PIK3CA or abnormal reduction of PTEN. Mutually exclusive alterations of PTEN and PIK3CA also suggest that mutations of either gene could activate the PI3-kinase/AKT signaling pathway, which is directly linked to the malignant progression of gastric tumor cells.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoric Monoester Hydrolases/genetics , Protein Serine-Threonine Kinases , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenoma/metabolism , Adenoma/pathology , DNA Methylation , DNA Mutational Analysis , DNA Primers/chemistry , DNA, Neoplasm/analysis , Gene Amplification , Gene Silencing , Hamartoma/genetics , Hamartoma/metabolism , Hamartoma/pathology , Humans , Loss of Heterozygosity , Male , Mutation , Neoplasm Staging , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pseudogenes/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...