Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 9(8): e106011, 2014.
Article in English | MEDLINE | ID: mdl-25162672

ABSTRACT

FOG1 is a transcriptional regulator that acts in concert with the hematopoietic master regulator GATA1 to coordinate the differentiation of platelets and erythrocytes. Despite considerable effort, however, the mechanisms through which FOG1 regulates gene expression are only partially understood. Here we report the discovery of a previously unrecognized domain in FOG1: a PR (PRD-BF1 and RIZ) domain that is distantly related in sequence to the SET domains that are found in many histone methyltransferases. We have used NMR spectroscopy to determine the solution structure of this domain, revealing that the domain shares close structural similarity with SET domains. Titration with S-adenosyl-L-homocysteine, the cofactor product synonymous with SET domain methyltransferase activity, indicated that the FOG PR domain is not, however, likely to function as a methyltransferase in the same fashion. We also sought to define the function of this domain using both pulldown experiments and gel shift assays. However, neither pulldowns from mammalian nuclear extracts nor yeast two-hybrid assays reproducibly revealed binding partners, and we were unable to detect nucleic-acid-binding activity in this domain using our high-diversity Pentaprobe oligonucleotides. Overall, our data demonstrate that FOG1 is a member of the PRDM (PR domain containing proteins, with zinc fingers) family of transcriptional regulators. The function of many PR domains, however, remains somewhat enigmatic for the time being.


Subject(s)
Nuclear Proteins/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Conserved Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Mice , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , S-Adenosylhomocysteine/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/genetics
2.
Nucleus ; 2(1): 30-7, 2011.
Article in English | MEDLINE | ID: mdl-21647297

ABSTRACT

A role for SUMOylation in the biogenesis and/or function of Box C/D snoRNPs has been reported, mediated via SUMO2 conjugation to the core snoRNP protein, Nop58. A quantitative proteomics screen, based on SILAC (stable-isotope labeling by amino acids in cell culture) and mass spectrometry using extracts prepared from cultured mammalian cells expressing either 6His-SUMO1 or -SUMO2, revealed that the snoRNP-related proteins Nop58, Nhp2, DKC1 and NOLC1 are amongst the main SUMO-modified proteins in the nucleolus. SUMOylation of Nhp2 and endogenous Nop58 was confirmed using a combination of in vitro and cell-based assays and the modified lysines identified by site-directed mutagenesis. SUMOylation of Nop58 was found to be important for high-affinity Box C/D snoRNA binding and the localization of newly transcribed snoRNAs to the nucleolus. Here, these findings are reviewed and a model for understanding Nop58 SUMOylation in the context of Box C/D snoRNP biogenesis is presented. Given the essential role of snoRNPs in the modification of pre-ribosomal RNA, this work suggests that SUMO, snoRNPs and ribosome assembly, and thus cellular translation, growth and proliferation, may be linked via novel mechanisms which warrant further investigation.


Subject(s)
Proteomics , Ribonucleoproteins, Small Nucleolar/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , HeLa Cells , Humans , Nuclear Proteins/metabolism , RNA, Small Nucleolar/metabolism , Ribonucleoproteins, Small Nuclear/metabolism
3.
Mol Cell ; 40(2): 216-27, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20965417

ABSTRACT

Cells typically respond quickly to stress, altering their metabolism to compensate. In mammalian cells, stress signaling usually leads to either cell-cycle arrest or apoptosis, depending on the severity of the insult and the ability of the cell to recover. Stress also often leads to reorganization of nuclear architecture, reflecting the simultaneous inhibition of major nuclear pathways (e.g., replication and transcription) and activation of specific stress responses (e.g., DNA repair). In this review, we focus on how two nuclear organelles, the nucleolus and the Cajal body, respond to stress. The nucleolus senses stress and is a central hub for coordinating the stress response. We review nucleolar function in the stress-induced regulation of p53 and the specific changes in nucleolar morphology and composition that occur upon stress. Crosstalk between nucleoli and CBs is also discussed in the context of stress responses.


Subject(s)
Cell Nucleolus/metabolism , Cell Nucleus/metabolism , Signal Transduction , Stress, Physiological/physiology , Animals , Coiled Bodies/metabolism , DNA Repair/physiology , Humans , Models, Biological , Tumor Suppressor Protein p53/physiology
4.
Proc Natl Acad Sci U S A ; 107(37): 16119-24, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805487

ABSTRACT

Protein modification by conjugation of small ubiquitin-related modifier (SUMO) is involved in diverse biological functions, such as transcription regulation, subcellular partitioning, stress response, DNA damage repair, and chromatin remodeling. Here, we show that the serine/arginine-rich protein SF2/ASF, a factor involved in splicing regulation and other RNA metabolism-related processes, is a regulator of the sumoylation pathway. The overexpression of this protein stimulates, but its knockdown inhibits SUMO conjugation. SF2/ASF interacts with Ubc9 and enhances sumoylation of specific substrates, sharing characteristics with already described SUMO E3 ligases. In addition, SF2/ASF interacts with the SUMO E3 ligase PIAS1 (protein inhibitor of activated STAT-1), regulating PIAS1-induced overall protein sumoylation. The RNA recognition motif 2 of SF2/ASF is necessary and sufficient for sumoylation enhancement. Moreover, SF2/ASF has a role in heat shock-induced sumoylation and promotes SUMO conjugation to RNA processing factors. These results add a component to the sumoylation pathway and a previously unexplored role for the multifunctional SR protein SF2/ASF.


Subject(s)
Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , SUMO-1 Protein/metabolism , Cell Line , Heat-Shock Response , Humans , Nuclear Proteins/genetics , Protein Binding , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors , Substrate Specificity , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
5.
Mol Cell ; 39(4): 618-31, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20797632

ABSTRACT

Posttranslational SUMO modification is an important mechanism of regulating protein function, especially in the cell nucleus. The nucleolus is the subnuclear organelle responsible for rRNA synthesis, processing, and assembly of the large and small ribosome subunits. Here, we have used SILAC-based quantitative proteomics to identify nucleolar SUMOylated proteins. This reveals a role for SUMOylation in the biogenesis and/or function of small nucleolar ribonucleoprotein complexes (snoRNPs) via the targeting of Nhp2 and Nop58. Using combined in vitro and in vivo approaches, both Nhp2 and Nop58 (also known as Nop5) are shown to be substrates for SUMOylation. Mutational analyses revealed the sites of modification on Nhp2 as K5, and on Nop58 as K467 and K497. Unlike Nop58 and Nhp2, the closely related Nop56 and 15.5K proteins appear not to be SUMO targets. SUMOylation is essential for high-affinity Nop58 binding to snoRNAs. This study provides direct evidence linking SUMO modification with snoRNP function.


Subject(s)
Cell Nucleolus/metabolism , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Proteomics , Ribonucleoproteins, Small Nucleolar/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Chromosomal Proteins, Non-Histone/metabolism , HeLa Cells , Humans , Lysine , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Proteomics/methods , Rats , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nucleolar/genetics , SUMO-1 Protein/metabolism , Transfection , Ubiquitins/metabolism
6.
Methods Enzymol ; 431: 83-111, 2007.
Article in English | MEDLINE | ID: mdl-17923232

ABSTRACT

MicroRNAs (miRs) are an important class of gene regulators that affect a wide range of biological processes. Despite the early recognition of miRs as translational regulators and intense interest in studying this phenomenon, it has so far not been possible to derive a consensus model for the underlying molecular mechanism(s). The potential of miRs to act in a combinatorial manner and to also promote mRNA decay creates conceptual and technical challenges in their study. Here, we discuss critical parameters in design and analysis of experiments used to study miR function including creation of synthetic miR and mRNA partners for assay of translational inhibition using luciferase reporters; measurement of mRNA stability after miR action; defining poly(A) tail length in miR target mRNA; determining the distribution of miRs and their target mRNAs in polysome profiles; and visualization of P-body components. We describe protocols for each of these procedures.


Subject(s)
Clinical Laboratory Techniques , Gene Expression Regulation , MicroRNAs/physiology , Protein Biosynthesis , Animals , Binding Sites , HeLa Cells , Humans , MicroRNAs/analysis , Plasmids , Polyadenylation , Polyribosomes/chemistry , RNA Stability , RNA, Messenger/genetics , Ribosomes/metabolism , Transfection
7.
J Mol Biol ; 362(2): 287-97, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16905148

ABSTRACT

The 22 kDa haem-binding protein, p22HBP, is highly expressed in erythropoietic tissues and binds to a range of metallo- and non-metalloporphyrin molecules with similar affinities, suggesting a role in haem regulation or synthesis. We have determined the three-dimensional solution structure of p22HBP and mapped the porphyrin-binding site, which comprises a number of loops and a alpha-helix all located on a single face of the molecule. The structure of p22HBP is related to the bacterial multi-drug resistance protein BmrR, and is the first protein with this fold to be identified in eukaryotes. Strikingly, the porphyrin-binding site in p22HBP is located in a similar position to the drug-binding site of BmrR. These similarities suggest that the broad ligand specificity observed for both BmrR and p22HBP may result from a conserved ligand interaction mechanism. Taken together, these data suggest that the both the fold and its associated function, that of binding to a broad range of small hydrophobic molecules, are ancient, and have been adapted throughout evolution for a variety of purposes.


Subject(s)
Carrier Proteins/chemistry , Heme/metabolism , Hemeproteins/chemistry , Porphyrins/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Circular Dichroism , Heme-Binding Proteins , Hemeproteins/genetics , Hemeproteins/metabolism , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Porphyrins/chemistry , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Trans-Activators/chemistry , Trans-Activators/metabolism
8.
Proc Natl Acad Sci U S A ; 102(47): 16961-6, 2005 Nov 22.
Article in English | MEDLINE | ID: mdl-16287976

ABSTRACT

MicroRNAs (miRNAs) repress translation of target mRNAs by interaction with partially mismatched sequences in their 3' UTR. The mechanism by which they act on translation has remained largely obscure. We examined the translation of mRNAs containing four partially mismatched miRNA-binding sites in the 3' UTR in HeLa cells cotransfected with a cognate miRNA. The mRNAs were prepared by in vitro transcription and were engineered to employ different modes of translation initiation. We find that the 5' cap structure and the 3' poly(A) tail are each necessary but not sufficient for full miRNA-mediated repression of mRNA translation. Replacing the cap structure with an internal ribosome entry site from either the cricket paralysis virus or the encephalomyocarditis virus impairs miRNA-mediated repression. Collectively, these results demonstrate that miRNAs interfere with the initiation step of translation and implicate the cap-binding protein eukaryotic initiation factor 4E as a molecular target.


Subject(s)
Eukaryotic Initiation Factor-4E/antagonists & inhibitors , MicroRNAs/physiology , Poly(A)-Binding Proteins/antagonists & inhibitors , RNA Caps/antagonists & inhibitors , HeLa Cells , Humans , RNA, Messenger/metabolism , Receptors, CXCR4/genetics , Ribosomes/physiology
9.
Biochemistry ; 43(42): 13318-27, 2004 Oct 26.
Article in English | MEDLINE | ID: mdl-15491138

ABSTRACT

The oligomerization domain that is present at the C terminus of Ikaros-family proteins and the protein Trps-1 is important for the proper regulation of developmental processes such as hematopoiesis. Remarkably, this domain is predicted to contain two classical zinc fingers (ZnFs), domains normally associated with the recognition of nucleic acids. The preference for protein binding by these predicted ZnFs is not well-understood. We have used a range of methods to gain insight into the structure of this domain. Circular dichroism, UV-vis, and NMR experiments carried out on the C-terminal domain of Eos (EosC) revealed that the two putative ZnFs (C1 and C2) are separable, i.e., capable of folding independently in the presence of Zn(II). We next determined the structure of EosC2 using NMR spectroscopy, revealing that, although the overall fold of EosC2 is similar to other classical ZnFs, a number of differences exist. For example, the conformation of the C terminus of EosC2 appears to be flexible and may result in a major rearrangement of the zinc ligands. Finally, alanine-scanning mutagenesis was used to identify the residues that are involved in the homo- and hetero-oligomerization of Eos, and these results are discussed in the context of the structure of EosC. These studies provide the first structural insights into how EosC mediates protein-protein interactions and contributes to our understanding of why it does not exhibit high-affinity DNA binding.


Subject(s)
Carrier Proteins/chemistry , DNA-Binding Proteins/chemistry , Nerve Tissue Proteins/chemistry , Peptide Fragments/chemistry , Transcription Factors/chemistry , Zinc Fingers , Amino Acid Sequence , Animals , Binding Sites/genetics , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Humans , Ikaros Transcription Factor , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nerve Tissue Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/genetics , Protein Binding/genetics , Protein Conformation , Protein Folding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary/genetics , Static Electricity , Structural Homology, Protein , Transcription Factors/genetics , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Zinc/chemistry , Zinc Fingers/genetics
10.
J Biol Chem ; 278(43): 42419-26, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-12917396

ABSTRACT

Ikaros family transcription factors play important roles in the control of hematopoiesis. Family members are predicted to contain up to six classic zinc fingers that are arranged into N- and C-terminal domains. The N-terminal domain is responsible for site-specific DNA binding, whereas the C-terminal domain primarily mediates the homo- and hetero-oligomerization between family members. Although the mechanisms of action of these proteins are not completely understood, the zinc finger domains are known to play a central role. In the current study, we have sought to understand the physical and functional properties of these domains, in particular the C-terminal domain. We show that the N-terminal domain from Eos, and not its C-terminal region, is required to recognize GGGA consensus sequences. Surprisingly, in contrast to the behavior exhibited by Ikaros, the C-terminal domain of Eos inhibits the DNA-binding activity of the full-length protein. In addition, we have used a range of biophysical techniques to demonstrate that the C-terminal domain of Eos mediates the formation of complexes that consist of nine or ten molecules. These results constitute the first direct demonstration that Ikaros family proteins can form higher order complexes in solution, and we discuss this unexpected result in the context of what is currently known about the family members and their possible mechanism of action.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/physiology , Consensus Sequence , Dimerization , Ikaros Transcription Factor , Nerve Tissue Proteins/physiology , Protein Binding , Protein Structure, Tertiary/physiology , Sequence Alignment , Solutions , Transcription Factors , Zinc Fingers/physiology
11.
J Biol Chem ; 278(25): 22805-11, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12657633

ABSTRACT

Identification of the protein domains that are responsible for RNA recognition has lagged behind the characterization of protein-DNA interactions. However, it is now becoming clear that a range of structural motifs bind to RNA and their structures and molecular mechanisms of action are beginning to be elucidated. In this report, we have expressed and purified one of the two putative RNA-binding domains from ZNF265, a protein that has been shown to bind to the spliceosomal components U1-70K and U2AF35 and to direct alternative splicing. We show that this domain, which contains four highly conserved cysteine residues, forms a stable, monomeric structure upon the addition of 1 molar eq of Zn(II). Determination of the solution structure of this domain reveals a conformation comprising two stacked beta-hairpins oriented at approximately 80 degrees to each other and sandwiching the zinc ion; the fold resembles the zinc ribbon class of zinc-binding domains, although with one less beta-strand than most members of the class. Analysis of the structure reveals a striking resemblance to known RNA-binding motifs in terms of the distribution of key surface residues responsible for making RNA contacts, despite a complete lack of structural homology. Furthermore, we have used an RNA gel shift assay to demonstrate that a single crossed finger domain from ZNF265 is capable of binding to an RNA message. Taken together, these results define a new RNA-binding motif and should provide insight into the functions of the >100 uncharacterized proteins in the sequence data bases that contain this domain.


Subject(s)
RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Zinc Fingers , Zinc/metabolism , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Conserved Sequence , Cysteine , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reproducibility of Results , Sequence Alignment , Sequence Homology, Amino Acid
12.
Int J Biochem Cell Biol ; 34(10): 1304-7, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12127581

ABSTRACT

Ikaros is an essential transcription factor for normal lymphocyte development. Because of its interaction with a number of closely related factors, Ikaros is required for correct regulation of differentiation and cell proliferation in T- and B-cell lineages. Interestingly, Ikaros appears to function both as a transcriptional repressor and as an activator through its ability to bind a large number of nuclear factors, including components of both histone deacetylase and ATP-dependent chromatin remodelling complexes. In addition, nuclear localisation is important for Ikaros function--unlike most transcription factors, Ikaros is localised to discrete nuclear foci in lymphoid cells, suggesting it employs novel mechanisms to regulate transcription.


Subject(s)
DNA-Binding Proteins , Hematopoiesis/physiology , Transcription Factors/physiology , Animals , Ikaros Transcription Factor , Protein Conformation , Transcription Factors/chemistry , Transcription Factors/genetics
13.
J Biol Chem ; 277(27): 24073-80, 2002 Jul 05.
Article in English | MEDLINE | ID: mdl-11940576

ABSTRACT

A number of structurally diverse classes of "antifreeze" proteins that allow fish to survive in sub-zero ice-laden waters have been isolated from the blood plasma of cold water teleosts. However, despite receiving a great deal of attention, the one or more mechanisms through which these proteins act are not fully understood. In this report we have synthesized a type I antifreeze polypeptide (AFP) from the shorthorn sculpin Myoxocephalus scorpius using recombinant methods. Construction of a synthetic gene with optimized codon usage and expression as a glutathione S-transferase fusion protein followed by purification yielded milligram amounts of polypeptide with two extra residues appended to the N terminus. Circular dichroism and NMR experiments, including residual dipolar coupling measurements on a 15N-labeled recombinant polypeptide, show that the polypeptides are alpha-helical with the first four residues being more flexible than the remainder of the sequence. Both the recombinant and synthetic polypeptides modify ice growth, forming facetted crystals just below the freezing point, but display negligible thermal hysteresis. Acetylation of Lys-10, Lys-20, and Lys-21 as well as the N terminus of the recombinant polypeptide gave a derivative that displays both thermal hysteresis (0.4 degrees C at 15 mg/ml) and ice crystal faceting. These results confirm that the N terminus of wild-type polypeptide is functionally important and support our previously proposed mechanism for all type I proteins, in which the hydrophobic face is oriented toward the ice at the ice/water interface.


Subject(s)
Antifreeze Proteins/chemistry , Fish Proteins , Ice , Amino Acid Sequence , Animals , Antifreeze Proteins/genetics , Base Sequence , Circular Dichroism , DNA Primers , Genes, Synthetic , Magnetic Resonance Spectroscopy , Microscopy, Video , Molecular Sequence Data , Protein Conformation , Recombinant Proteins/biosynthesis , Scorpions , Solutions
SELECTION OF CITATIONS
SEARCH DETAIL
...