Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Cell ; 165(3): 606-19, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27104978

ABSTRACT

Rbfox proteins control alternative splicing and posttranscriptional regulation in mammalian brain and are implicated in neurological disease. These proteins recognize the RNA sequence (U)GCAUG, but their structures and diverse roles imply a variety of protein-protein interactions. We find that nuclear Rbfox proteins are bound within a large assembly of splicing regulators (LASR), a multimeric complex containing the proteins hnRNP M, hnRNP H, hnRNP C, Matrin3, NF110/NFAR-2, NF45, and DDX5, all approximately equimolar to Rbfox. We show that splicing repression mediated by hnRNP M is stimulated by Rbfox. Virtually all the intron-bound Rbfox is associated with LASR, and hnRNP M motifs are enriched adjacent to Rbfox crosslinking sites in vivo. These findings demonstrate that Rbfox proteins bind RNA with a defined set of cofactors and affect a broader set of exons than previously recognized. The function of this multimeric LASR complex has implications for deciphering the regulatory codes controlling splicing networks.


Subject(s)
RNA Splicing , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Animals , Brain/cytology , Brain/metabolism , Cell Nucleus/metabolism , Exons , HEK293 Cells , Humans , Introns , Mice , Multiprotein Complexes/metabolism , RNA Precursors/metabolism
2.
Cell ; 166(4): 1041-1054, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27499020

ABSTRACT

We used clinical tissue from lethal metastatic castration-resistant prostate cancer (CRPC) patients obtained at rapid autopsy to evaluate diverse genomic, transcriptomic, and phosphoproteomic datasets for pathway analysis. Using Tied Diffusion through Interacting Events (TieDIE), we integrated differentially expressed master transcriptional regulators, functionally mutated genes, and differentially activated kinases in CRPC tissues to synthesize a robust signaling network consisting of druggable kinase pathways. Using MSigDB hallmark gene sets, six major signaling pathways with phosphorylation of several key residues were significantly enriched in CRPC tumors after incorporation of phosphoproteomic data. Individual autopsy profiles developed using these hallmarks revealed clinically relevant pathway information potentially suitable for patient stratification and targeted therapies in late stage prostate cancer. Here, we describe phosphorylation-based cancer hallmarks using integrated personalized signatures (pCHIPS) that shed light on the diversity of activated signaling pathways in metastatic CRPC while providing an integrative, pathway-based reference for drug prioritization in individual patients.


Subject(s)
Phosphoproteins/analysis , Prostatic Neoplasms, Castration-Resistant/chemistry , Proteome/analysis , Algorithms , Humans , Male , Precision Medicine , Prostatic Neoplasms, Castration-Resistant/metabolism , Signal Transduction , Transcriptome
3.
Cell ; 157(5): 1050-60, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24855943

ABSTRACT

DNA methylation is a conserved epigenetic gene-regulation mechanism. DOMAINS REARRANGED METHYLTRANSFERASE (DRM) is a key de novo methyltransferase in plants, but how DRM acts mechanistically is poorly understood. Here, we report the crystal structure of the methyltransferase domain of tobacco DRM (NtDRM) and reveal a molecular basis for its rearranged structure. NtDRM forms a functional homodimer critical for catalytic activity. We also show that Arabidopsis DRM2 exists in complex with the small interfering RNA (siRNA) effector ARGONAUTE4 (AGO4) and preferentially methylates one DNA strand, likely the strand acting as the template for RNA polymerase V-mediated noncoding RNA transcripts. This strand-biased DNA methylation is also positively correlated with strand-biased siRNA accumulation. These data suggest a model in which DRM2 is guided to target loci by AGO4-siRNA and involves base-pairing of associated siRNAs with nascent RNA transcripts.


Subject(s)
Arabidopsis/enzymology , Methyltransferases/metabolism , Nicotiana/enzymology , Amino Acid Sequence , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Argonaute Proteins/metabolism , Catalytic Domain , Methyltransferases/chemistry , Models, Molecular , Molecular Sequence Data , Nicotiana/metabolism
4.
Cell ; 151(1): 167-80, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-23021223

ABSTRACT

DNA methylation and histone modification exert epigenetic control over gene expression. CHG methylation by CHROMOMETHYLASE3 (CMT3) depends on histone H3K9 dimethylation (H3K9me2), but the mechanism underlying this relationship is poorly understood. Here, we report multiple lines of evidence that CMT3 interacts with H3K9me2-containing nucleosomes. CMT3 genome locations nearly perfectly correlated with H3K9me2, and CMT3 stably associated with H3K9me2-containing nucleosomes. Crystal structures of maize CMT3 homolog ZMET2, in complex with H3K9me2 peptides, showed that ZMET2 binds H3K9me2 via both bromo adjacent homology (BAH) and chromo domains. The structures reveal an aromatic cage within both BAH and chromo domains as interaction interfaces that capture H3K9me2. Mutations that abolish either interaction disrupt CMT3 binding to nucleosomes and show a complete loss of CMT3 activity in vivo. Our study establishes dual recognition of H3K9me2 marks by BAH and chromo domains and reveals a distinct mechanism of interplay between DNA methylation and histone modification.


Subject(s)
Arabidopsis/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA, Plant/metabolism , Nucleosomes/metabolism , Zea mays/metabolism , Amino Acid Sequence , Arabidopsis/genetics , Crystallography, X-Ray , DNA (Cytosine-5-)-Methyltransferases/chemistry , Heterochromatin/metabolism , Histones/metabolism , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Zea mays/genetics
5.
Mol Cell ; 75(2): 382-393.e5, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31229404

ABSTRACT

The iron-sensing protein FBXL5 is the substrate adaptor for a SKP1-CUL1-RBX1 E3 ubiquitin ligase complex that regulates the degradation of iron regulatory proteins (IRPs). Here, we describe a mechanism of FBXL5 regulation involving its interaction with the cytosolic Fe-S cluster assembly (CIA) targeting complex composed of MMS19, FAM96B, and CIAO1. We demonstrate that the CIA-targeting complex promotes the ability of FBXL5 to degrade IRPs. In addition, the FBXL5-CIA-targeting complex interaction is regulated by oxygen (O2) tension displaying a robust association in 21% O2 that is severely diminished in 1% O2 and contributes to O2-dependent regulation of IRP degradation. Together, these data identify a novel oxygen-dependent signaling axis that links IRP-dependent iron homeostasis with the Fe-S cluster assembly machinery.


Subject(s)
Cell Cycle Proteins/genetics , F-Box Proteins/genetics , Molecular Chaperones/genetics , Multiprotein Complexes/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Cell Cycle Proteins/chemistry , F-Box Proteins/chemistry , HeLa Cells , Homeostasis , Humans , Iron/metabolism , Iron-Regulatory Proteins/genetics , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Molecular Chaperones/chemistry , Multiprotein Complexes/chemistry , Oxygen/metabolism , Proteolysis , Transcription Factors/genetics , Ubiquitin-Protein Ligase Complexes/chemistry
6.
J Biol Chem ; 298(7): 102094, 2022 07.
Article in English | MEDLINE | ID: mdl-35654137

ABSTRACT

The cytosolic iron-sulfur (Fe-S) cluster assembly (CIA) pathway delivers Fe-S clusters to nuclear and cytosolic Fe-S proteins involved in essential cellular functions. Although the delivery process is regulated by the availability of iron and oxygen, it remains unclear how CIA components orchestrate the cluster transfer under varying cellular environments. Here, we utilized a targeted proteomics assay for monitoring CIA factors and substrates to characterize the CIA machinery. We find that nucleotide-binding protein 1 (NUBP1/NBP35), cytosolic iron-sulfur assembly component 3 (CIAO3/NARFL), and CIA substrates associate with nucleotide-binding protein 2 (NUBP2/CFD1), a component of the CIA scaffold complex. NUBP2 also weakly associates with the CIA targeting complex (MMS19, CIAO1, and CIAO2B) indicating the possible existence of a higher order complex. Interactions between CIAO3 and the CIA scaffold complex are strengthened upon iron supplementation or low oxygen tension, while iron chelation and reactive oxygen species weaken CIAO3 interactions with CIA components. We further demonstrate that CIAO3 mutants defective in Fe-S cluster binding fail to integrate into the higher order complexes. However, these mutants exhibit stronger associations with CIA substrates under conditions in which the association with the CIA targeting complex is reduced suggesting that CIAO3 and CIA substrates may associate in complexes independently of the CIA targeting complex. Together, our data suggest that CIA components potentially form a metabolon whose assembly is regulated by environmental cues and requires Fe-S cluster incorporation in CIAO3. These findings provide additional evidence that the CIA pathway adapts to changes in cellular environment through complex reorganization.


Subject(s)
Iron-Sulfur Proteins , Iron , Cytosol/metabolism , GTP-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Iron/metabolism , Iron-Sulfur Proteins/biosynthesis , Iron-Sulfur Proteins/metabolism , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Sulfur/metabolism
7.
Mol Cell ; 55(3): 495-504, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25018018

ABSTRACT

In Arabidopsis, CHG DNA methylation is controlled by the H3K9 methylation mark through a self-reinforcing loop between DNA methyltransferase CHROMOMETHYLASE3 (CMT3) and H3K9 histone methyltransferase KRYPTONITE/SUVH4 (KYP). We report on the structure of KYP in complex with methylated DNA, substrate H3 peptide, and cofactor SAH, thereby defining the spatial positioning of the SRA domain relative to the SET domain. The methylated DNA is bound by the SRA domain with the 5mC flipped out of the DNA, while the H3(1-15) peptide substrate binds between the SET and post-SET domains, with the ε-ammonium of K9 positioned adjacent to bound SAH. These structural insights, complemented by functional data on key mutants of residues lining the 5mC and H3K9-binding pockets within KYP, establish how methylated DNA recruits KYP to the histone substrate. Together, the structures of KYP and previously reported CMT3 complexes provide insights into molecular mechanisms linking DNA and histone methylation.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/genetics , DNA Methylation , DNA, Plant/chemistry , DNA, Plant/genetics , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Histones/physiology , Arabidopsis/chemistry , Arabidopsis/metabolism , Binding Sites/genetics , Epigenesis, Genetic , Gene Expression Regulation, Plant , Genome, Plant , Models, Molecular , S-Adenosylhomocysteine/metabolism , X-Ray Diffraction
8.
J Proteome Res ; 20(9): 4318-4330, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34342229

ABSTRACT

G-protein-coupled receptors (GPCRs) initiate intracellular signaling events through heterotrimeric G-protein α-subunits (Gα) and the ßγ-subunit dimer (Gßγ). In this study, we utilized mass spectrometry to identify novel regulators of Gßγ signaling in human cells. This prompted our characterization of KCTD2 and KCTD5, two related potassium channel tetramerization domain (KCTD) proteins that specifically recognize Gßγ. We demonstrated that these KCTD proteins are substrate adaptors for a multisubunit CUL3-RING ubiquitin ligase, in which a KCTD2-KCTD5 hetero-oligomer associates with CUL3 through KCTD5 subunits and recruits Gßγ through both KCTD proteins in response to G-protein activation. These KCTD proteins promote monoubiquitination of lysine-23 within Gß1/2in vitro and in HEK-293 cells. Depletion of these adaptors from cancer cell lines sharply impairs downstream signaling. Together, our studies suggest that a KCTD2-KCTD5-CUL3-RING E3 ligase recruits Gßγ in response to signaling, monoubiquitinates lysine-23 within Gß1/2, and regulates Gßγ effectors to modulate downstream signal transduction.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Ubiquitin-Protein Ligases , Cullin Proteins/genetics , Cullin Proteins/metabolism , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Potassium Channels , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
Mol Cell ; 51(1): 105-15, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23747013

ABSTRACT

Zinc is an essential cofactor of all major eukaryotic RNA polymerases. How the activity of these enzymes is coordinated or regulated according to cellular zinc levels is largely unknown. Here we show that the stability of RNA polymerase I (RNAPI) is tightly coupled to zinc availability in vivo. In zinc deficiency, RNAPI is specifically degraded by proteolysis in the vacuole in a pathway dependent on the export in Xpo1p and deubiquitination of the RNAPI large subunit Rpa190p by Ubp2p and Ubp4p. RNAPII is unaffected, which allows for the expression of genes required in zinc deficiency. RNAPI export to the vacuole is required for survival during zinc starvation, suggesting that degradation of zinc-binding subunits might provide a last resort zinc reservoir. These results reveal a hierarchy of cellular transcriptional activities during zinc starvation and show that degradation of the most active cellular transcriptional machinery couples cellular growth and proliferation to zinc availability.


Subject(s)
RNA Polymerase I/physiology , Saccharomyces cerevisiae/growth & development , Zinc/metabolism , Down-Regulation , Endopeptidases/metabolism , Endopeptidases/physiology , Enzyme Stability , RNA Polymerase I/metabolism , RNA, Ribosomal/biosynthesis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Ubiquitination , Vacuoles/metabolism
10.
Mol Cell Proteomics ; 17(11): 2229-2241, 2018 11.
Article in English | MEDLINE | ID: mdl-29444981

ABSTRACT

The flagellated protozoan parasite Trichomonas vaginalis is the etiologic agent of trichomoniasis, the most common non-viral sexually transmitted infection worldwide. As an obligate extracellular pathogen, adherence to epithelial cells is critical for parasite survival within the human host and a better understanding of this process is a prerequisite for the development of therapies to combat infection. In this sense, recent work has shown S-acylation as a key modification that regulates pathogenesis in different protozoan parasites. However, there are no reports indicating whether this post-translational modification is a mechanism operating in T. vaginalis In order to study the extent and function of S-acylation in T. vaginalis biology, we undertook a proteomic study to profile the full scope of S-acylated proteins in this parasite and reported the identification of 363 proteins involved in a variety of biological processes such as protein transport, pathogenesis related and signaling, among others. Importantly, treatment of parasites with the palmitoylation inhibitor 2-bromopalmitate causes a significant decrease in parasite: parasite aggregation as well as adherence to host cells suggesting that palmitoylation could be modifying proteins that are key regulators of Trichomonas vaginalis pathogenesis.


Subject(s)
Lipoylation , Protozoan Proteins/metabolism , Trichomonas vaginalis/metabolism , Adhesiveness , Amino Acid Sequence , Gene Ontology , HeLa Cells , Humans , Protein Domains , Proteome/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/isolation & purification
11.
Cell Mol Life Sci ; 75(12): 2211-2226, 2018 06.
Article in English | MEDLINE | ID: mdl-29222644

ABSTRACT

Trichomonas vaginalis is a common sexually transmitted parasite that colonizes the human urogenital tract, where it remains extracellular and adheres to epithelial cells. Infections range from asymptomatic to highly inflammatory, depending on the host and the parasite strain. Despite the serious consequences associated with trichomoniasis disease, little is known about parasite or host factors involved in attachment of the parasite-to-host epithelial cells. Here, we report the identification of microvesicle-like structures (MVs) released by T. vaginalis. MVs are considered universal transport vehicles for intercellular communication as they can incorporate peptides, proteins, lipids, miRNA, and mRNA, all of which can be transferred to target cells through receptor-ligand interactions, fusion with the cell membrane, and delivery of a functional cargo to the cytoplasm of the target cell. In the present study, we demonstrated that T. vaginalis release MVs from the plasma and the flagellar membranes of the parasite. We performed proteomic profiling of these structures demonstrating that they possess physical characteristics similar to mammalian extracellular vesicles and might be selectively charged with specific protein content. In addition, we demonstrated that viable T. vaginalis parasites release large vesicles (LVs), membrane structures larger than 1 µm that are able to interact with other parasites and with the host cell. Finally, we show that both populations of vesicles present on the surface of T vaginalis are induced in the presence of host cells, consistent with a role in modulating cell interactions.


Subject(s)
Extracellular Vesicles/metabolism , Host-Parasite Interactions , Trichomonas Vaginitis/metabolism , Trichomonas Vaginitis/parasitology , Trichomonas vaginalis/physiology , Trichomonas vaginalis/ultrastructure , Cell Communication , Extracellular Vesicles/chemistry , Extracellular Vesicles/ultrastructure , Female , HeLa Cells , Humans , Proteomics , Protozoan Proteins/analysis , Protozoan Proteins/metabolism , Trichomonas vaginalis/chemistry , Trichomonas vaginalis/cytology
12.
Nucleic Acids Res ; 45(4): 2081-2098, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28204614

ABSTRACT

MicroRNAs (miRNAs) impinge on the translation and stability of their target mRNAs, and play key roles in development, homeostasis and disease. The gene regulation mechanisms they instigate are largely mediated through the CCR4­NOT deadenylase complex, but the molecular events that occur on target mRNAs are poorly resolved. We observed a broad convergence of interactions of germ granule and P body mRNP components on AIN-1/GW182 and NTL-1/CNOT1 in Caenorhabditis elegans embryos. We show that the miRISC progressively matures on the target mRNA from a scanning form into an effector mRNP particle by sequentially recruiting the CCR4­NOT complex, decapping and decay, or germ granule proteins. Finally, we implicate intrinsically disordered proteins, key components in mRNP architectures, in the embryonic function of lsy-6 miRNA. Our findings define dynamic steps of effector mRNP assembly in miRNA-mediated silencing, and identify a functional continuum between germ granules and P bodies in the C. elegans embryo.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/metabolism , RNA Interference , Ribonucleoproteins/metabolism , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cytoplasmic Granules/metabolism , Embryo, Nonmammalian/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intrinsically Disordered Proteins/metabolism , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/metabolism , Ribonucleases/metabolism
13.
Proc Natl Acad Sci U S A ; 113(3): 632-7, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26721397

ABSTRACT

Cilia (eukaryotic flagella) are present in diverse eukaryotic lineages and have essential motility and sensory functions. The cilium's capacity to sense and transduce extracellular signals depends on dynamic trafficking of ciliary membrane proteins. This trafficking is often mediated by the Bardet-Biedl Syndrome complex (BBSome), a protein complex for which the precise subcellular distribution and mechanisms of action are unclear. In humans, BBSome defects perturb ciliary membrane protein distribution and manifest clinically as Bardet-Biedl Syndrome. Cilia are also important in several parasites that cause tremendous human suffering worldwide, yet biology of the parasite BBSome remains largely unexplored. We examined BBSome functions in Trypanosoma brucei, a flagellated protozoan parasite that causes African sleeping sickness in humans. We report that T. brucei BBS proteins assemble into a BBSome that interacts with clathrin and is localized to membranes of the flagellar pocket and adjacent cytoplasmic vesicles. Using BBS gene knockouts and a mouse infection model, we show the T. brucei BBSome is dispensable for flagellar assembly, motility, bulk endocytosis, and cell viability but required for parasite virulence. Quantitative proteomics reveal alterations in the parasite surface proteome of BBSome mutants, suggesting that virulence defects are caused by failure to maintain fidelity of the host-parasite interface. Interestingly, among proteins altered are those with ubiquitination-dependent localization, and we find that the BBSome interacts with ubiquitin. Collectively, our data indicate that the BBSome facilitates endocytic sorting of select membrane proteins at the base of the cilium, illuminating BBSome roles at a critical host-pathogen interface and offering insights into BBSome molecular mechanisms.


Subject(s)
Bardet-Biedl Syndrome/metabolism , Endocytosis , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei brucei/pathogenicity , Animals , Clathrin/metabolism , Flagella/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Mice , Parasites/pathogenicity , Protein Binding , Protein Transport , Transport Vesicles/metabolism , Virulence
14.
PLoS Genet ; 12(5): e1005998, 2016 05.
Article in English | MEDLINE | ID: mdl-27171361

ABSTRACT

The MORC family of GHKL ATPases are an enigmatic class of proteins with diverse chromatin related functions. In Arabidopsis, AtMORC1, AtMORC2, and AtMORC6 act together in heterodimeric complexes to mediate transcriptional silencing of methylated DNA elements. Here, we studied Arabidopsis AtMORC4 and AtMORC7. We found that, in contrast to AtMORC1,2,6, they act to suppress a wide set of non-methylated protein-coding genes that are enriched for those involved in pathogen response. Furthermore, atmorc4 atmorc7 double mutants show a pathogen response phenotype. We found that AtMORC4 and AtMORC7 form homomeric complexes in vivo and are concentrated in discrete nuclear bodies adjacent to chromocenters. Analysis of an atmorc1,2,4,5,6,7 hextuple mutant demonstrates that transcriptional de-repression is largely uncoupled from changes in DNA methylation in plants devoid of MORC function. However, we also uncover a requirement for MORC in both DNA methylation and silencing at a small but distinct subset of RNA-directed DNA methylation target loci. These regions are characterized by poised transcriptional potential and a low density of sites for symmetric cytosine methylation. These results provide insight into the biological function of MORC proteins in higher eukaryotes.


Subject(s)
Adenosine Triphosphatases/genetics , Arabidopsis Proteins/genetics , Epigenesis, Genetic , Transcription, Genetic , Adenosine Triphosphatases/biosynthesis , Arabidopsis/genetics , Arabidopsis Proteins/biosynthesis , DNA Methylation/genetics , Gene Expression Regulation, Plant , Multigene Family/genetics , Phenotype
15.
Genes Dev ; 25(20): 2198-209, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21979373

ABSTRACT

Murine Chd1 (chromodomain helicase DNA-binding protein 1), a chromodomain-containing chromatin remodeling protein, is necessary for embryonic stem (ES) cell pluripotency. Chd1 binds to nucleosomes trimethylated at histone 3 Lys 4 (H3K4me3) near the beginning of active genes but not to bivalent domains also containing H3K27me3. To address the mechanism of this specificity, we reproduced H3K4me3- and CHD1-stimulated gene activation in HeLa extracts. Multidimensional protein identification technology (MuDPIT) and immunoblot analyses of purified preinitiation complexes (PICs) revealed the recruitment of CHD1 to naive chromatin but enhancement on H3K4me3 chromatin. Studies in depleted extracts showed that the Mediator coactivator complex, which controls PIC assembly, is also necessary for CHD1 recruitment. MuDPIT analyses of CHD1-associated proteins support the recruitment data and reveal numerous components of the PIC, including Mediator. In vivo, CHD1 and Mediator are recruited to an inducible gene, and genome-wide binding of the two proteins correlates well with active gene transcription in mouse ES cells. Finally, coimmunoprecipitation of CHD1 and Mediator from cell extracts can be ablated by shRNA knockdown of a specific Mediator subunit. Our data support a model in which the Mediator coordinates PIC assembly along with the recruitment of CHD1. The combined action of the PIC and H3K4me3 provides specificity in targeting CHD1 to active genes.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Mediator Complex/metabolism , Animals , Gene Expression Regulation , HeLa Cells , Histones/metabolism , Humans , Immunoprecipitation , Mediator Complex/genetics , Mice , Protein Binding , Proteomics
16.
Curr Genet ; 64(4): 821-839, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29288414

ABSTRACT

Trypanosomatids are parasitic protozoans characterized by several unique structural and metabolic processes that include exquisite mechanisms associated with gene expression and regulation. During the initiation of protein synthesis, for instance, mRNA selection for translation seems to be mediated by different eIF4F-like complexes, which may play a significant role in parasite adaptation to different hosts. In eukaryotes, the heterotrimeric eIF4F complex (formed by eIF4E, eIF4G, and eIF4A) mediates mRNA recognition and ribosome binding and participates in various translation regulatory events. Six eIF4Es and five eIF4Gs have been described in trypanosomatids with several of these forming different eIF4F-like complexes. This has raised questions about their role in differential mRNA translation. Here we have studied further TbEIF4E2, the least known eIF4E homologue from Trypanosoma brucei, and found that it is not associated with an eIF4G homolog. It is, however, associated with mature mRNAs and binds to a histone mRNA stem-loop-binding protein (SLBP), one of two Trypanosoma SLBP homologs (TbSLBP1 and TbSLBP2). TbSLBP1 is more similar to the mammalian counterpart while TbSLBP2 is exclusive to trypanosomatids and related organisms. TbSLBP2 binds to TbEIF4E2 through a conserved central region missing in other SLBP homologs. Both SLBPs, as well as TbEIF4E2, were found to localize to the cytoplasm. TbEIF4E2 and TbSLBP2 are differentially expressed during cell culture, being more abundant in early-log phase, with TbSLBP2 also showing cell-cycle dependent expression. The new data reinforce unique aspects of the trypanosomatid eIF4Es, with the TbEIF4E2-TbSLBP complex possibly having a role in differential selection of mRNAs containing stem-loop structures.


Subject(s)
Eukaryotic Initiation Factor-4E/genetics , Nuclear Proteins/genetics , Trypanosoma brucei brucei/genetics , Trypanosomiasis/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Amino Acid Sequence/genetics , Gene Expression/genetics , Histones/genetics , Humans , Protein Binding , Protein Biosynthesis/genetics , RNA Cap-Binding Proteins/genetics , RNA, Messenger/genetics , Sequence Alignment , Trypanosomiasis/parasitology
17.
Genes Cells ; 22(10): 850-860, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28771906

ABSTRACT

In eukaryotic cells, a surveillance mechanism, the S phase checkpoint, detects and responds to insults that challenge chromosomal replication, arresting cell cycle progression and triggering appropriate events to prevent genomic instability. In the budding yeast Saccharomyces cerevisiae, Mec1/ATM/ATR, and its downstream kinase, Rad53/Chk2, mediate the response to genotoxic stress. In this study, we place Cip1, a recently identified Cdk1 inhibitor (CKI), under the regulation of Mec1 and Rad53 in response to genotoxic stress. Cip1 accumulates dramatically in a Mec1- and Rad53-dependent manner upon replication stress. This increase requires the activity of MBF, but not the transcriptional activator kinase Dun1. At the protein level, stabilization of replication stress-induced Cip1 requires continued de novo protein synthesis. In addition, Cip1 is phosphorylated at an S/TQ motif in a Mec1-dependent manner. Deletion of Cip1 affects proliferation in hydroxyurea-containing plates. Significantly, the sensitivity is increased when the dosage of the G1 cyclin CLN2 is increased, compatible to a role of Cip1 as a G1-cyclin-dependent kinase inhibitor. In all, our results place Cip1 under the S phase checkpoint response to genotoxic stress. Furthermore, Cip1 plays a significant role to preserve viability in response to insults that threaten chromosome replication.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , S Phase , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Amino Acid Motifs , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Checkpoint Kinase 2/metabolism , Cyclins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
18.
Cell Microbiol ; 19(4)2017 04.
Article in English | MEDLINE | ID: mdl-27696623

ABSTRACT

The Toxoplasma inner membrane complex (IMC) is a specialized organelle underlying the parasite's plasma membrane that consists of flattened rectangular membrane sacs that are sutured together and positioned atop a supportive cytoskeleton. We have previously identified a novel class of proteins localizing to the transverse and longitudinal sutures of the IMC, which we named IMC sutures components (ISCs). Here, we have used proximity-dependent biotin identification at the sutures to better define the composition of this IMC subcompartment. Using ISC4 as bait, we demonstrate biotin-dependent labeling of the sutures and have uncovered two new ISCs. We also identified five new proteins that exclusively localize to the transverse sutures that we named transverse sutures components (TSCs), demonstrating that components of the IMC sutures consist of two groups: those that localize to the transverse and longitudinal sutures (ISCs) and those residing only in the transverse sutures (TSCs). In addition, we functionally analyze the ISC protein ISC3 and demonstrate that ISC3-null parasites have morphological defects and reduced fitness in vitro. Most importantly, Δisc3 parasites exhibit a complete loss of virulence in vivo. These studies expand the known composition of the IMC sutures and highlight the contribution of ISCs to the ability of the parasite to proliferate and cause disease.


Subject(s)
Protozoan Proteins/physiology , Toxoplasma/ultrastructure , Cells, Cultured , Female , Gene Knockout Techniques , Host-Parasite Interactions , Humans , Phosphatidate Phosphatase/physiology , Phosphatidate Phosphatase/ultrastructure , Protozoan Proteins/ultrastructure , Toxoplasma/physiology , Virulence
19.
Mol Cell ; 40(4): 558-70, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21095586

ABSTRACT

To understand how miRNA-mediated silencing impacts on embryonic mRNAs, we conducted a functional survey of abundant maternal and zygotic miRNA families in the C. elegans embryo. We show that the miR-35-42 and the miR-51-56 miRNA families define maternal and zygotic miRNA-induced silencing complexes (miRISCs), respectively, that share a large number of components. Using a cell-free C. elegans embryonic extract, we demonstrate that the miRISC directs the rapid deadenylation of reporter mRNAs with natural 3'UTRs. The deadenylated targets are translationally suppressed and remarkably stable. Sampling of the predicted miR-35-42 targets reveals that roughly half are deadenylated in a miRNA-dependent manner, but with each target displaying a distinct efficiency and pattern of deadenylation. Finally, we demonstrate that functional cooperation between distinct miRISCs within 3'UTRs is required to potentiate deadenylation. With this report, we reveal the extensive and direct impact of miRNA-mediated deadenylation on embryonic mRNAs.


Subject(s)
3' Untranslated Regions/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Embryo, Nonmammalian/metabolism , MicroRNAs/genetics , RNA 3' End Processing , Animals , Base Sequence , Cell-Free System , Female , Gene Silencing , MicroRNAs/metabolism , Models, Biological , Molecular Sequence Data , Proteomics , RNA-Induced Silencing Complex/metabolism , Zygote/metabolism
20.
PLoS Genet ; 11(9): e1005468, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26332045

ABSTRACT

A surveillance mechanism, the S phase checkpoint, blocks progression into mitosis in response to DNA damage and replication stress. Segregation of damaged or incompletely replicated chromosomes results in genomic instability. In humans, the S phase checkpoint has been shown to constitute an anti-cancer barrier. Inhibition of mitotic cyclin dependent kinase (M-CDK) activity by Wee1 kinases is critical to block mitosis in some organisms. However, such mechanism is dispensable in the response to genotoxic stress in the model eukaryotic organism Saccharomyces cerevisiae. We show here that the Wee1 ortholog Swe1 does indeed inhibit M-CDK activity and chromosome segregation in response to genotoxic insults. Swe1 dispensability in budding yeast is the result of a redundant control of M-CDK activity by the checkpoint kinase Rad53. In addition, our results indicate that Swe1 is an effector of the checkpoint central kinase Mec1. When checkpoint control on M-CDK and on Pds1/securin stabilization are abrogated, cells undergo aberrant chromosome segregation.


Subject(s)
Chromosome Segregation , Chromosomes, Fungal , DNA Damage , DNA Replication , Saccharomyces cerevisiae/genetics , Cyclin-Dependent Kinases/metabolism , Mutagens/toxicity , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL