Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
PLoS Genet ; 18(7): e1010305, 2022 07.
Article in English | MEDLINE | ID: mdl-35789210

ABSTRACT

Circadian clocks enable organisms to predict and align their behaviors and physiologies to constant daily day-night environmental cycle. Because the ubiquitin ligase Siah2 has been identified as a potential regulator of circadian clock function in cultured cells, we have used SIAH2-deficient mice to examine its function in vivo. Our experiments demonstrate a striking and unexpected sexually dimorphic effect of SIAH2-deficiency on the regulation of rhythmically expressed genes in the liver. The absence of SIAH2 in females, but not in males, altered the expression of core circadian clock genes and drastically remodeled the rhythmic transcriptome in the liver by increasing the number of day-time expressed genes, and flipping the rhythmic expression from nighttime expressed genes to the daytime. These effects are not readily explained by effects on known sexually dimorphic pathways in females. Moreover, loss of SIAH2 in females, not males, preferentially altered the expression of transcription factors and genes involved in regulating lipid and lipoprotein metabolism. Consequently, SIAH2-deficient females, but not males, displayed disrupted daily lipid and lipoprotein patterns, increased adiposity and impaired metabolic homeostasis. Overall, these data suggest that SIAH2 may be a key component of a female-specific circadian transcriptional output circuit that directs the circadian timing of gene expression to regulate physiological rhythms, at least in the liver. In turn, our findings imply that sex-specific transcriptional mechanisms may closely interact with the circadian clock to tailor overt rhythms for sex-specific needs.


Subject(s)
Circadian Clocks , Circadian Rhythm , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Female , Lipids , Lipoproteins , Male , Mice , Ubiquitin , Ubiquitin-Protein Ligases/genetics
2.
Hum Mol Genet ; 28(1): 16-30, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30215740

ABSTRACT

Polycystin-1 (PC1), encoded by the PKD1 gene that is mutated in the autosomal dominant polycystic kidney disease, regulates a number of processes including bone development. Activity of the transcription factor RunX2, which controls osteoblast differentiation, is reduced in Pkd1 mutant mice but the mechanism governing PC1 activation of RunX2 is unclear. PC1 undergoes regulated cleavage that releases its C-terminal tail (CTT), which translocates to the nucleus to modulate transcriptional pathways involved in proliferation and apoptosis. We find that the cleaved CTT of PC1 (PC1-CTT) stimulates the transcriptional coactivator TAZ (Wwtr1), an essential coactivator of RunX2. PC1-CTT physically interacts with TAZ, stimulating RunX2 transcriptional activity in pre-osteoblast cells in a TAZ-dependent manner. The PC1-CTT increases the interaction between TAZ and RunX2 and enhances the recruitment of the p300 transcriptional co-regulatory protein to the TAZ/RunX2/PC1-CTT complex. Zebrafish injected with morpholinos directed against pkd1 manifest severe bone calcification defects and a curly tail phenotype. Injection of messenger RNA (mRNA) encoding the PC1-CTT into pkd1-morphant fish restores bone mineralization and reduces the severity of the curly tail phenotype. These effects are abolished by co-injection of morpholinos directed against TAZ. Injection of mRNA encoding a dominant-active TAZ construct is sufficient to rescue both the curly tail phenotype and the skeletal defects observed in pkd1-morpholino treated fish. Thus, TAZ constitutes a key mechanistic link through which PC1 mediates its physiological functions.


Subject(s)
Bone Development/genetics , Intracellular Signaling Peptides and Proteins/physiology , TRPP Cation Channels/physiology , Animals , Apoptosis , Bone Development/physiology , Cell Differentiation , E1A-Associated p300 Protein/physiology , Gene Expression Regulation , Genes, Regulator , HEK293 Cells , Humans , Kidney/metabolism , Models, Animal , Morpholinos , Osteoblasts/metabolism , Osteogenesis/physiology , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
J Biol Chem ; 293(15): 5478-5491, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29475951

ABSTRACT

Syntaxins are a conserved family of SNARE proteins and contain C-terminal transmembrane anchors required for their membrane fusion activity. Here we show that Stx3 (syntaxin 3) unexpectedly also functions as a nuclear regulator of gene expression. We found that alternative splicing creates a soluble isoform that we termed Stx3S, lacking the transmembrane anchor. Soluble Stx3S binds to the nuclear import factor RanBP5 (RAN-binding protein 5), targets to the nucleus, and interacts physically and functionally with several transcription factors, including ETV4 (ETS variant 4) and ATF2 (activating transcription factor 2). Stx3S is differentially expressed in normal human tissues, during epithelial cell polarization, and in breast cancer versus normal breast tissue. Inhibition of endogenous Stx3S expression alters the expression of cancer-associated genes and promotes cell proliferation. Similar nuclear-targeted, soluble forms of other syntaxins were identified, suggesting that nuclear signaling is a conserved, novel function common among these membrane-trafficking proteins.


Subject(s)
Adenovirus E1A Proteins/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Gene Expression Regulation , Proto-Oncogene Proteins/metabolism , Qa-SNARE Proteins/metabolism , Signal Transduction , beta Karyopherins/metabolism , Adenovirus E1A Proteins/genetics , Animals , COS Cells , Caco-2 Cells , Cell Nucleus/genetics , Chlorocebus aethiops , Dogs , HEK293 Cells , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets , Qa-SNARE Proteins/genetics , Solubility , beta Karyopherins/genetics
4.
PLoS Biol ; 13(7): e1002200, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26196739

ABSTRACT

The protozoan parasite, Toxoplasma, like many intracellular pathogens, suppresses interferon gamma (IFN-γ)-induced signal transducer and activator of transcription 1 (STAT1) activity. We exploited this well-defined host-pathogen interaction as the basis for a high-throughput screen, identifying nine transcription factors that enhance STAT1 function in the nucleus, including the orphan nuclear hormone receptor TLX. Expression profiling revealed that upon IFN-γ treatment TLX enhances the output of a subset of IFN-γ target genes, which we found is dependent on TLX binding at those loci. Moreover, infection of TLX deficient mice with the intracellular parasite Toxoplasma results in impaired production of the STAT1-dependent cytokine interleukin-12 by dendritic cells and increased parasite burden in the brain during chronic infection. These results demonstrate a previously unrecognized role for this orphan nuclear hormone receptor in regulating STAT1 signaling and host defense and reveal that STAT1 activity can be modulated in a context-specific manner by such "modifiers."


Subject(s)
Gene Expression Regulation , Host-Pathogen Interactions , Receptors, Cytoplasmic and Nuclear/metabolism , STAT1 Transcription Factor/metabolism , Toxoplasma/immunology , Animals , Brain/metabolism , Central Nervous System Infections/metabolism , Chemokine CXCL10/metabolism , Chemokine CXCL9/metabolism , Chronic Disease , Interferon-gamma/metabolism , Mice, Inbred CBA
5.
Proc Natl Acad Sci U S A ; 112(40): 12420-5, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26392558

ABSTRACT

Regulated degradation of proteins by the proteasome is often critical to their function in dynamic cellular pathways. The molecular clock underlying mammalian circadian rhythms relies on the rhythmic expression and degradation of its core components. However, because the tools available for identifying the mechanisms underlying the degradation of a specific protein are limited, the mechanisms regulating clock protein degradation are only beginning to be elucidated. Here we describe a cell-based functional screening approach designed to quickly identify the ubiquitin E3 ligases that induce the degradation of potentially any protein of interest. We screened the nuclear hormone receptor RevErbα (Nr1d1), a key constituent of the mammalian circadian clock, for E3 ligases that regulate its stability and found Seven in absentia2 (Siah2) to be a key regulator of RevErbα stability. Previously implicated in hypoxia signaling, Siah2 overexpression destabilizes RevErbα/ß, and siRNA depletion of Siah2 stabilizes endogenous RevErbα. Moreover, Siah2 depletion delays circadian degradation of RevErbα and lengthens period length. These results demonstrate the utility of functional screening approaches for identifying regulators of protein stability and reveal Siah2 as a previously unidentified circadian clockwork regulator that mediates circadian RevErbα turnover.


Subject(s)
Circadian Clocks/genetics , Nuclear Proteins/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Expression , Humans , Mice , Microscopy, Fluorescence , Nuclear Proteins/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Proteolysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Protein Ligases/metabolism
6.
PLoS Biol ; 12(4): e1001840, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24737000

ABSTRACT

Over the last decades, researchers have characterized a set of "clock genes" that drive daily rhythms in physiology and behavior. This arduous work has yielded results with far-reaching consequences in metabolic, psychiatric, and neoplastic disorders. Recent attempts to expand our understanding of circadian regulation have moved beyond the mutagenesis screens that identified the first clock components, employing higher throughput genomic and proteomic techniques. In order to further accelerate clock gene discovery, we utilized a computer-assisted approach to identify and prioritize candidate clock components. We used a simple form of probabilistic machine learning to integrate biologically relevant, genome-scale data and ranked genes on their similarity to known clock components. We then used a secondary experimental screen to characterize the top candidates. We found that several physically interact with known clock components in a mammalian two-hybrid screen and modulate in vitro cellular rhythms in an immortalized mouse fibroblast line (NIH 3T3). One candidate, Gene Model 129, interacts with BMAL1 and functionally represses the key driver of molecular rhythms, the BMAL1/CLOCK transcriptional complex. Given these results, we have renamed the gene CHRONO (computationally highlighted repressor of the network oscillator). Bi-molecular fluorescence complementation and co-immunoprecipitation demonstrate that CHRONO represses by abrogating the binding of BMAL1 to its transcriptional co-activator CBP. Most importantly, CHRONO knockout mice display a prolonged free-running circadian period similar to, or more drastic than, six other clock components. We conclude that CHRONO is a functional clock component providing a new layer of control on circadian molecular dynamics.


Subject(s)
ARNTL Transcription Factors/metabolism , Circadian Clocks/physiology , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Artificial Intelligence , Cell Line , Circadian Clocks/genetics , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Circadian Rhythm Signaling Peptides and Proteins/biosynthesis , Circadian Rhythm Signaling Peptides and Proteins/genetics , Cryptochromes/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Glucocorticoid/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Sequence Alignment , Transcription, Genetic/genetics
7.
Proc Natl Acad Sci U S A ; 109(38): 15348-53, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22949635

ABSTRACT

Endoderm-mesenchyme cross-talk is a central process in the development of foregut-derived organs. How signaling pathways integrate the activity of multiple ligands to guide organ development is poorly understood. We show that two Wnt ligands, Wnt2 and Wnt7b, cooperatively induce Wnt signaling without affecting the stabilization of the Wnt canonical effector ß-catenin despite it being necessary for Wnt2-Wnt7b cooperativity. Wnt2-Wnt7b cooperation is specific for mesenchymal cell lineages and the combined loss of Wnt2 and Wnt7b leads to more severe developmental defects in the lung than loss of Wnt2 or Wnt7b alone. High-throughput small-molecule screens and biochemical assays reveal that the Pdgf pathway is required for cooperative Wnt2-Wnt7b signaling. Inhibition of Pdgf signaling in cell culture reduces Wnt2-Wnt7b cooperative signaling. Moreover, inhibition of Pdgf signaling in lung explant cultures results in decreased Wnt signaling and lung smooth-muscle development. These data suggest a model in which Pdgf signaling potentiates Wnt2-Wnt7b signaling to promote high levels of Wnt activity in mesenchymal progenitors that is required for proper development of endoderm-derived organs, such as the lung.


Subject(s)
Gene Expression Regulation, Developmental , Intestinal Mucosa/metabolism , Intestines/embryology , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins/metabolism , Wnt Proteins/metabolism , Wnt2 Protein/metabolism , Animals , Cell Line , Cell Lineage , Epithelium/metabolism , Humans , Ligands , Lung/metabolism , Mesoderm/metabolism , Mice , Myocytes, Smooth Muscle/metabolism , Organogenesis/genetics , Signal Transduction
8.
Nat Genet ; 38(3): 312-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16474406

ABSTRACT

Direct evidence for the requirement of transcriptional feedback repression in circadian clock function has been elusive. Here, we developed a molecular genetic screen in mammalian cells to identify mutants of the circadian transcriptional activators CLOCK and BMAL1, which were uncoupled from CRYPTOCHROME (CRY)-mediated transcriptional repression. Notably, mutations in the PER-ARNT-SIM domain of CLOCK and the C terminus of BMAL1 resulted in synergistic insensitivity through reduced physical interactions with CRY. Coexpression of these mutant proteins in cultured fibroblasts caused arrhythmic phenotypes in population and single-cell assays. These data demonstrate that CRY-mediated repression of the CLOCK/BMAL1 complex activity is required for maintenance of circadian rhythmicity and provide formal proof that transcriptional feedback is required for mammalian clock function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Circadian Rhythm/physiology , Gene Expression Regulation , Trans-Activators/genetics , 3T3 Cells , ARNTL Transcription Factors , Animals , CLOCK Proteins , Cell Line , Feedback , Genes, Reporter , Humans , Luciferases/analysis , Luciferases/genetics , Luminescence , Mice , Plasmids , Time
9.
Annu Rev Pharmacol Toxicol ; 50: 187-214, 2010.
Article in English | MEDLINE | ID: mdl-20055702

ABSTRACT

The physiology of a wide variety of organisms is organized according to periodic environmental changes imposed by the earth's rotation. This way, a large number of physiological processes present diurnal rhythms regulated by an internal timing system called the circadian clock. As part of the rhythmicity in physiology, drug efficacy and toxicity can vary with time. Studies over the past four decades present diurnal oscillations in drug absorption, distribution, metabolism, and excretion. On the other hand, diurnal variations in the availability and sensitivity of drug targets have been correlated with time-dependent changes in drug effectiveness. In this review, we provide evidence supporting the regulation of drug kinetics and dynamics by the circadian clock. We also use the examples of hypertension and cancer to show current achievements and challenges in chronopharmacology.


Subject(s)
Biological Clocks/genetics , Circadian Rhythm , Pharmacokinetics , Pharmacology , ARNTL Transcription Factors/genetics , Animals , Blood Pressure , CLOCK Proteins/genetics , Cyclic GMP/biosynthesis , Humans , Hypertension/drug therapy , Neoplasms/drug therapy , Nitric Oxide/biosynthesis , Renin-Angiotensin System/drug effects , Suprachiasmatic Nucleus/physiology
10.
PLoS Biol ; 7(3): e52, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19278294

ABSTRACT

The mammalian circadian clock is a cell-autonomous system that drives oscillations in behavior and physiology in anticipation of daily environmental change. To assess the robustness of a human molecular clock, we systematically depleted known clock components and observed that circadian oscillations are maintained over a wide range of disruptions. We developed a novel strategy termed Gene Dosage Network Analysis (GDNA) in which small interfering RNA (siRNA)-induced dose-dependent changes in gene expression were used to build gene association networks consistent with known biochemical constraints. The use of multiple doses powered the analysis to uncover several novel network features of the circadian clock, including proportional responses and signal propagation through interacting genetic modules. We also observed several examples where a gene is up-regulated following knockdown of its paralog, suggesting the clock network utilizes active compensatory mechanisms rather than simple redundancy to confer robustness and maintain function. We propose that these network features act in concert as a genetic buffering system to maintain clock function in the face of genetic and environmental perturbation.


Subject(s)
Biological Clocks/genetics , Circadian Rhythm/genetics , Gene Expression Regulation , Gene Regulatory Networks , Mammals/genetics , Animals , Gene Duplication , Gene Knockdown Techniques , Humans , Mammals/physiology , Mice , Mice, Knockout , Models, Animal , Models, Genetic , RNA, Small Interfering , Signal Transduction
11.
PLoS Genet ; 5(4): e1000442, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19343201

ABSTRACT

The circadian clock is a molecular and cellular oscillator found in most mammalian tissues that regulates rhythmic physiology and behavior. Numerous investigations have addressed the contribution of circadian rhythmicity to cellular, organ, and organismal physiology. We recently developed a method to look at transcriptional oscillations with unprecedented precision and accuracy using high-density time sampling. Here, we report a comparison of oscillating transcription from mouse liver, NIH3T3, and U2OS cells. Several surprising observations resulted from this study, including a 100-fold difference in the number of cycling transcripts in autonomous cellular models of the oscillator versus tissues harvested from intact mice. Strikingly, we found two clusters of genes that cycle at the second and third harmonic of circadian rhythmicity in liver, but not cultured cells. Validation experiments show that 12-hour oscillatory transcripts occur in several other peripheral tissues as well including heart, kidney, and lungs. These harmonics are lost ex vivo, as well as under restricted feeding conditions. Taken in sum, these studies illustrate the importance of time sampling with respect to multiple testing, suggest caution in use of autonomous cellular models to study clock output, and demonstrate the existence of harmonics of circadian gene expression in the mouse.


Subject(s)
Circadian Rhythm , Liver/physiology , Mammals/genetics , Transcription, Genetic , Animals , Cell Line , Cells, Cultured , Gene Expression , Humans , Male , Mammals/physiology , Mice , Mice, Inbred C57BL , NIH 3T3 Cells
12.
Bioinformatics ; 24(23): 2794-5, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18931366

ABSTRACT

UNLABELLED: Oscillations in mRNA and protein of circadian clock components can be continuously monitored in vitro using synchronized cell lines. These rhythms can be highly variable due to culture conditions and are non-stationary due to baseline trends, damping and drift in period length. We present a technique for characterizing the modal frequencies of oscillation using continuous wavelet decomposition to non-parametrically model changes in amplitude and period while removing baseline effects and noise. AVAILABILITY: The method has been implemented as the package waveclock for the free statistical software program R and is available for download from http://cran.r-project.org/


Subject(s)
Circadian Rhythm/physiology , Computational Biology/methods , Algorithms , Animals , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
13.
Curr Opin Genet Dev ; 15(6): 634-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16226457

ABSTRACT

Comparative genomics approaches are having a remarkable impact on the study of transcriptional regulation in eukaryotes. Many eukaryotic genome sequences are being explored by new computational methods and high-throughput experimental tools such as DNA arrays and genome-wide location analyses. These tools are enabling efficient panning for common regulatory cassettes underlying fundamental biological processes, extending the use of existing techniques for the discovery of response elements to mammals, deciphering the transcriptional regulatory code in eukaryotes and providing the first global insights into a recently described post-transcriptional regulatory mechanism. Collectively, these approaches are rapidly expanding both our knowledge and our definition of transcriptional regulation.


Subject(s)
Eukaryotic Cells/physiology , Gene Expression Regulation/genetics , Genomics , Transcription, Genetic/genetics , Animals , Gene Expression Regulation/physiology , Humans , Transcription, Genetic/physiology
14.
Curr Biol ; 13(3): 189-98, 2003 Feb 04.
Article in English | MEDLINE | ID: mdl-12573214

ABSTRACT

BACKGROUND: Different types of regulation are utilized to produce a robust circadian clock, including regulation at the transcriptional, posttranscriptional, and translational levels. A screen for rhythmic messages that may be involved in such circadian control identified nocturnin, a novel gene that displays high-amplitude circadian expression in the Xenopus laevis retina, with peak mRNA levels in the early night. Expression of nocturnin mRNA is confined to the clock-containing photoreceptor cell layer within the retina. RESULTS: In these studies, we show that nocturnin removes the poly(A) tail from a synthetic RNA substrate in a process known as deadenylation. Nocturnin nuclease activity is magnesium dependent, as the addition of EDTA or mutation of the residue predicted to bind magnesium disrupts deadenylation. Substrate preference studies show that nocturnin is an exonuclease that specifically degrades the 3' poly(A) tail. While nocturnin is rhythmically expressed in the cytoplasm of the retinal photoreceptor cells, the only other described vertebrate deadenylase, PARN, is constitutively present in most retinal cells, including the photoreceptors. CONCLUSIONS: The distinct spatial and temporal expression of nocturnin and PARN suggests that there may be specific mRNA targets of each deadenylase. Since deadenylation regulates mRNA decay and/or translational silencing, we propose that nocturnin deadenylates clock-related transcripts in a novel mechanism for posttranscriptional regulation in the circadian clock or its outputs.


Subject(s)
Circadian Rhythm/physiology , Gene Expression Regulation , Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Retina/enzymology , Animals , Biological Clocks/physiology , Exoribonucleases/genetics , Exoribonucleases/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Humans , Magnesium/metabolism , Nuclear Proteins , Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/metabolism , Transcription Factors , Xenopus laevis
15.
Methods Mol Biol ; 317: 243-54, 2006.
Article in English | MEDLINE | ID: mdl-16264233

ABSTRACT

Within the retina there is a circadian clock that controls the 24-h timing of processes such as hormone release, cell movement, and gene transcription. In an effort to better understand the molecular nature of this retinal clock, a differential display (DD) screen was performed to isolate a gene with high amplitude circadian rhythmicity in the Xenopus retina. A novel gene expressed in the early evening in photoreceptor cells was isolated and named nocturnin for night factor. This article outlines the steps we took to study a protein of unknown function, particularly highlighting the analyses one can perform when little more than the primary sequence of a gene is known. In addition, we describe the results of sequence analysis that assisted in predicting the function of nocturnin. We have shown that nocturnin acts as a deadenylase in vitro, removing the poly(A) tail from a mature messenger RNA in a process that either leads to degradation or translational silencing of a message. Although the role of nocturnin in the retina is unknown, future studies to identify target mRNAs that are deadenylated by nocturnin will assist in elucidating its physiological role in this tissue.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation , Proteins/genetics , Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Circadian Rhythm , Edetic Acid/chemistry , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Nuclear Proteins , Promoter Regions, Genetic , Protein Structure, Tertiary , Proteins/chemistry , RNA, Messenger/metabolism , Retina/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors , Transcription, Genetic , Xenopus
16.
Cell Rep ; 9(5): 1885-1895, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25466249

ABSTRACT

Loss of Pax3, a developmentally regulated transcription factor expressed in premigratory neural crest, results in severe developmental defects and embryonic lethality. Although Pax3 mutations produce profound phenotypes, the intrinsic transcriptional activation exhibited by Pax3 is surprisingly modest. We postulated the existence of transcriptional coactivators that function with Pax3 to mediate developmental functions. A high-throughput screen identified the Hippo effector proteins Taz and Yap65 as Pax3 coactivators. Synergistic coactivation of target genes by Pax3-Taz/Yap65 requires DNA binding by Pax3, is Tead independent, and is regulated by Hippo kinases Mst1 and Lats2. In vivo, Pax3 and Yap65 colocalize in the nucleus of neural crest progenitors in the dorsal neural tube. Neural crest deletion of Taz and Yap65 results in embryo-lethal neural crest defects and decreased expression of the Pax3 target gene, Mitf. These results suggest that Pax3 activity is regulated by the Hippo pathway and that Pax factors are Hippo effectors.


Subject(s)
Gene Expression Regulation, Developmental , Melanocytes/metabolism , Neural Crest/cytology , Paired Box Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Genes, Reporter , HEK293 Cells , Hippo Signaling Pathway , Humans , Luciferases/biosynthesis , Luciferases/genetics , Mice, Transgenic , PAX3 Transcription Factor , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Transcriptional Activation
17.
PLoS One ; 8(1): e55782, 2013.
Article in English | MEDLINE | ID: mdl-23383281

ABSTRACT

Previous studies have demonstrated that certain Wnt ligands can promote high levels of cooperative signaling in a cell type specific manner. To explore the underlying mechanism of this cooperative Wnt signaling, we performed a high-throughput screen of more than 14,000 cDNAs to identify genes that promote cooperative Wnt signaling in the context of a single Wnt ligand, Wnt2. This screen identified several homeobox factors including Msx2, Nkx5.2, and Esx1, in addition to other factors known to promote Wnt signaling including Pias4. Generation of dominant-active or dominant-negative forms of Msx2 indicate that the mechanism by which homeobox factors cooperatively promote Wnt signaling is through their ability to repress gene transcription. These data identify a broad homeobox code, which acts to increase Wnt signaling through transcriptional repression.


Subject(s)
Genomics , Wnt Proteins/metabolism , Wnt Signaling Pathway , Cell Line , Gene Expression Profiling , Gene Expression Regulation , Genomics/methods , High-Throughput Screening Assays , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Ligands , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
18.
Dev Cell ; 22(1): 197-210, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22178500

ABSTRACT

Mutations in Pkd1, encoding polycystin-1 (PC1), cause autosomal-dominant polycystic kidney disease (ADPKD). We show that the carboxy-terminal tail (CTT) of PC1 is released by γ-secretase-mediated cleavage and regulates the Wnt and CHOP pathways by binding the transcription factors TCF and CHOP, disrupting their interaction with the common transcriptional coactivator p300. Loss of PC1 causes increased proliferation and apoptosis, while reintroducing PC1-CTT into cultured Pkd1 null cells reestablishes normal growth rate, suppresses apoptosis, and prevents cyst formation. Inhibition of γ-secretase activity impairs the ability of PC1 to suppress growth and apoptosis and leads to cyst formation in cultured renal epithelial cells. Expression of the PC1-CTT is sufficient to rescue the dorsal body curvature phenotype in zebrafish embryos resulting from either γ-secretase inhibition or suppression of Pkd1 expression. Thus, γ-secretase-dependent release of the PC1-CTT creates a protein fragment whose expression is sufficient to suppress ADPKD-related phenotypes in vitro and in vivo.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Apoptosis , TCF Transcription Factors/metabolism , TRPP Cation Channels/physiology , Transcription Factor CHOP/metabolism , Zebrafish/metabolism , p300-CBP Transcription Factors/genetics , Animals , Cell Proliferation , Cells, Cultured , Cysts/etiology , Cysts/metabolism , Cysts/pathology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Humans , Immunoblotting , Immunoprecipitation , Kidney/metabolism , Kidney/pathology , Phenotype , Polycystic Kidney, Autosomal Dominant/physiopathology , TCF Transcription Factors/genetics , TRPP Cation Channels/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcriptional Activation , Wnt Signaling Pathway , Zebrafish/genetics , Zebrafish/growth & development , p300-CBP Transcription Factors/metabolism
19.
Curr Opin Genet Dev ; 20(6): 581-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926286

ABSTRACT

The circadian clock is an endogenous oscillator that regulates daily rhythms in behavior and physiology. In recent years, systems biology and genomics approaches re-shaped our view of the clock. Our understanding of outputs that regulate behavior and physiology has been enhanced through gene expression profiling and proteomic analyses. Systems approaches uncovered underlying principles of transcriptional regulation and robustness of the oscillator through perturbation analysis and synthetic methods. Finally, new clock components and modifiers were identified through cell-based screening efforts and proteomics.


Subject(s)
Circadian Rhythm , Genomics , Systems Biology , Animals , Humans
20.
Article in English | MEDLINE | ID: mdl-20836017

ABSTRACT

The conventional target centric model of drug discovery is pinned under the weight of prior success and the traditional problems of safety and efficacy for new molecules. An alternative to target centric drug development is to shift focus to the pathways that mediate both biology and pathophysiology. This method has the advantage of not requiring a priori knowledge of the small molecule target, but also comes with it several challenges including target determination. We suggest extending this notion more broadly across the drug discovery process using quantitative network structure-activity relationships (QNSAR), and discuss the steps necessary to test the hypothesis that systems biology approaches can be used to improve the drug discovery process.


Subject(s)
Drug Discovery , Systems Biology , Algorithms , Chemistry, Pharmaceutical/methods , Genome, Human , Genomics , Humans , Proteomics , Quantitative Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL