Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Traffic ; 20(12): 890-911, 2019 12.
Article in English | MEDLINE | ID: mdl-31606941

ABSTRACT

The intracellular environment is partitioned into functionally distinct compartments containing specific sets of molecules and reactions. Biomolecular condensates, also referred to as membrane-less organelles, are diverse and abundant cellular compartments that lack membranous enclosures. Molecules assemble into condensates by phase separation; multivalent weak interactions drive molecules to separate from their surroundings and concentrate in discrete locations. Biomolecular condensates exist in all eukaryotes and in some prokaryotes, and participate in various essential house-keeping, stress-response and cell type-specific processes. An increasing number of recent studies link abnormal condensate formation, composition and material properties to a number of disease states. In this review, we discuss current knowledge and models describing the regulation of condensates and how they become dysregulated in neurodegeneration and cancer. Further research on the regulation of biomolecular phase separation will help us to better understand their role in cell physiology and disease.


Subject(s)
Cytoplasmic Granules/metabolism , Neoplasms/metabolism , Animals , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/metabolism , Cytoplasmic Granules/chemistry , Cytoplasmic Granules/pathology , Humans , Neoplasms/pathology
2.
Semin Cell Dev Biol ; 90: 94-103, 2019 06.
Article in English | MEDLINE | ID: mdl-30017905

ABSTRACT

Proteins and RNAs inside the cell nucleus are organized into distinct phases, also known as liquid-liquid phase separated (LLPS) droplet organelles or nuclear bodies. These regions exist within the spaces between chromatin-rich regions but their function is tightly linked to gene activity. They include major microscopically-observable structures such as the nucleolus, paraspeckle and Cajal body. The biochemical and assembly factors enriched inside these microenvironments regulate chromatin structure, transcription, and RNA processing, and other important cellular functions. Here, we describe published evidence that suggests nuclear bodies are bona fide LLPS droplet organelles and major regulators of the processes listed above. We also outline an updated "Supply or Sequester" model to describe nuclear body function, in which proteins or RNAs are supplied to surrounding genomic regions or sequestered away from their sites of activity. Finally, we describe recent evidence that suggests these microenvironments are both reflective and drivers of diverse pathophysiological states.


Subject(s)
Cell Nucleus Structures/metabolism , Cell Nucleus/chemistry , Cell Separation , Epigenesis, Genetic/genetics , Liquid-Liquid Extraction , Organelles/metabolism , RNA/metabolism , Cell Nucleus/metabolism , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/genetics , Humans , Organelles/chemistry , Organelles/genetics , Particle Size , RNA/genetics , RNA/isolation & purification
3.
Nat Rev Mol Cell Biol ; 8(12): 1006-16, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17928811

ABSTRACT

The promyelocytic leukaemia (PML) tumour suppressor protein epitomizes the PML-nuclear body (PML-NB) and is crucially required for the proper assembly of this macromolecular nuclear structure. Unlike other, more specialized subnuclear structures such as Cajal and Polycomb group bodies, PML-NBs are functionally promiscuous and have been implicated in the regulation of diverse cellular functions. PML-NBs are dynamic structures that favour the sequestration and release of proteins, mediate their post-translational modifications and promote specific nuclear events in response to various cellular stresses. Recent data suggest that PML-NBs may be heterogeneous in composition, mobility and function.


Subject(s)
Intranuclear Inclusion Bodies/chemistry , Intranuclear Inclusion Bodies/physiology , Animals , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/physiology , Humans , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
4.
Biochem Biophys Res Commun ; 483(1): 142-146, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28039056

ABSTRACT

The loop domain organization of chromatin plays an important role in transcription regulation and thus may be assumed to vary in cells of different types. We investigated the kinetics of DNA loop migration during single cell gel electrophoresis (the comet assay) for nucleoids obtained from human lymphocytes, lymphoblasts and glioblastoma T98G cells. The results confirm our previous observation that there are three parts of DNA in nucleoids: DNA on the nucleoid surface, loops up to ∼150 kb inside the nucleoid, and larger loops that cannot migrate. However, the relative amounts of the three parts were found to be very different for different cell types. The distributions of the loop length up to 150 kb were shown to be exponential, with the distribution parameter, the loop density, to be dependent on the cell type.


Subject(s)
Comet Assay/methods , DNA/chemistry , Adult , Cell Nucleus Structures/chemistry , Female , Humans , Kinetics , Lymphocytes/cytology , Lymphocytes/physiology , Male
5.
Nucleic Acids Res ; 43(9): 4661-75, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25870410

ABSTRACT

Single genes are often subject to alternative splicing, which generates alternative mature mRNAs. This phenomenon is widespread in animals, and observed in over 90% of human genes. Recent data suggest it may also be common in Apicomplexa. These parasites have small genomes, and economy of DNA is evolutionarily favoured in this phylum. We investigated the mechanism of alternative splicing in Toxoplasma gondii, and have identified and localized TgSR3, a homologue of ASF/SF2 (alternative-splicing factor/splicing factor 2, a serine-arginine-rich, or SR protein) to a subnuclear compartment. In addition, we conditionally overexpressed this protein, which was deleterious to growth. qRT-PCR was used to confirm perturbation of splicing in a known alternatively-spliced gene. We performed high-throughput RNA-seq to determine the extent of splicing modulated by this protein. Current RNA-seq algorithms are poorly suited to compact parasite genomes, and hence we complemented existing tools by writing a new program, GeneGuillotine, that addresses this deficiency by segregating overlapping reads into distinct genes. In order to identify the extent of alternative splicing, we released another program, JunctionJuror, that detects changes in intron junctions. Using this program, we identified about 2000 genes that were constitutively alternatively spliced in T. gondii. Overexpressing the splice regulator TgSR3 perturbed alternative splicing in over 1000 genes.


Subject(s)
Alternative Splicing , Nuclear Proteins/metabolism , Protozoan Proteins/metabolism , RNA-Binding Proteins/metabolism , Toxoplasma/genetics , Cell Nucleus Structures/chemistry , Gene Expression , Nuclear Proteins/analysis , Nuclear Proteins/classification , Nuclear Proteins/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/analysis , Protozoan Proteins/classification , Protozoan Proteins/genetics , RNA-Binding Proteins/analysis , RNA-Binding Proteins/classification , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors , Software , Toxoplasma/growth & development , Toxoplasma/metabolism
6.
Nucleic Acids Res ; 42(13): 8648-62, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25013176

ABSTRACT

Nuclear paraspeckles are built co-transcriptionally around a long non-coding RNA, NEAT1. Here we report that transfected 20-mer phosphorothioate-modified (PS) antisense oligonucleotides (ASOs) can recruit paraspeckle proteins to form morphologically normal and apparently functional paraspeckle-like structures containing no NEAT1 RNA. PS-ASOs can associate with paraspeckle proteins, including P54nrb, PSF, PSPC1 and hnRNPK. NEAT1 RNA can be displaced by transfected PS-ASO from paraspeckles and rapidly degraded. Co-localization of PS-ASOs with P54nrb was observed in canonical NEAT1-containing paraspeckles, in perinucleolar caps upon transcriptional inhibition, and importantly, in paraspeckle-like or filament structures lacking NEAT1 RNA. The induced formation of paraspeckle-like and filament structures occurred in mouse embryonic stem cells expressing little or no NEAT1 RNA, suggesting that PS-ASOs can serve as seeding molecules to assemble paraspeckle-like foci in the absence of NEAT1 RNA. Moreover, CTN, an RNA reported to be functionally retained in paraspeckles, was also observed to localize to paraspeckle-like structures, implying that paraspeckle-like structures assembled on PS-ASOs are functional. Together, our results indicate that functional paraspeckles can form with short nucleic acids other than NEAT1 RNA.


Subject(s)
Cell Nucleus Structures/chemistry , Oligonucleotides, Antisense/analysis , Phosphorothioate Oligonucleotides/analysis , RNA, Long Noncoding/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , HeLa Cells , Humans , Mice , Nuclear Proteins/analysis , Oligonucleotides, Antisense/metabolism , Phosphorothioate Oligonucleotides/metabolism , Transfection
7.
Nucleic Acids Res ; 42(22): 13788-98, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25414336

ABSTRACT

Mammalian splicing regulatory protein RNA-binding motif protein 4 (RBM4) has an alanine repeat-containing C-terminal domain (CAD) that confers both nuclear- and splicing speckle-targeting activities. Alanine-repeat expansion has pathological potential. Here we show that the alanine-repeat tracts influence the subnuclear targeting properties of the RBM4 CAD in cultured human cells. Notably, truncation of the alanine tracts redistributed a portion of RBM4 to paraspeckles. The alanine-deficient CAD was sufficient for paraspeckle targeting. On the other hand, alanine-repeat expansion reduced the mobility of RBM4 and impaired its splicing activity. We further took advantage of the putative coactivator activator (CoAA)-RBM4 conjoined splicing factor, CoAZ, to investigate the function of the CAD in subnuclear targeting. Transiently expressed CoAZ formed discrete nuclear foci that emerged and subsequently separated-fully or partially-from paraspeckles. Alanine-repeat expansion appeared to prevent CoAZ separation from paraspeckles, resulting in their complete colocalization. CoAZ foci were dynamic but, unlike paraspeckles, were resistant to RNase treatment. Our results indicate that the alanine-rich CAD, in conjunction with its conjoined RNA-binding domain(s), differentially influences the subnuclear localization and biogenesis of RBM4 and CoAZ.


Subject(s)
Alanine , Alternative Splicing , Cell Nucleus Structures/chemistry , Nuclear Proteins/chemistry , Peptides , Protein Sorting Signals , RNA-Binding Proteins/chemistry , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism , Repetitive Sequences, Amino Acid
8.
Nucleic Acids Res ; 42(12): 7819-32, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24861627

ABSTRACT

Phosphorothioate (PS) antisense oligonucleotides (ASOs) have been successfully developed as drugs to reduce the expression of disease-causing genes. PS-ASOs can be designed to induce degradation of complementary RNAs via the RNase H pathway and much is understood about that process. However, interactions of PS-ASOs with other cellular proteins are not well characterized. Here we report that in cells transfected with PS-ASOs, the chaperonin T-complex 1 (TCP1) proteins interact with PS-ASOs and enhance antisense activity. The TCP1-ß subunit co-localizes with PS-ASOs in distinct nuclear structures, termed phosphorothioate bodies or PS-bodies. Upon Ras-related nuclear protein (RAN) depletion, cytoplasmic PS-body-like structures were observed and nuclear concentrations of PS-ASOs were reduced, suggesting that TCP1-ß can interact with PS-ASOs in the cytoplasm and that the nuclear import of PS-ASOs is at least partially through the RAN-mediated pathway. Upon free uptake, PS-ASOs co-localize with TCP1 proteins in cytoplasmic foci related to endosomes/lysosomes. Together, our results indicate that the TCP1 complex binds oligonucleotides with TCP1-ß subunit being a nuclear PS-body component and suggest that the TCP1 complex may facilitate PS-ASO uptake and/or release from the endocytosis pathway.


Subject(s)
Cell Nucleus Structures/chemistry , Chaperonin Containing TCP-1/metabolism , Oligonucleotides, Antisense/metabolism , Phosphorothioate Oligonucleotides/metabolism , Cell Nucleus/metabolism , Chaperonin Containing TCP-1/analysis , Chaperonin Containing TCP-1/isolation & purification , Cytoplasm/chemistry , Endocytosis , HeLa Cells , Humans , Oligonucleotides, Antisense/analysis , Oligonucleotides, Antisense/isolation & purification , Phosphorothioate Oligonucleotides/analysis , Phosphorothioate Oligonucleotides/isolation & purification , Protein Subunits/analysis , Transfection , ran GTP-Binding Protein/antagonists & inhibitors
9.
Nucleic Acids Res ; 41(8): 4447-58, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23444137

ABSTRACT

We have previously shown that α-thalassemia mental retardation X-linked (ATRX) and histone H3.3 are key regulators of telomeric chromatin in mouse embryonic stem cells. The function of ATRX and H3.3 in the maintenance of telomere chromatin integrity is further demonstrated by recent studies that show the strong association of ATRX/H3.3 mutations with alternative lengthening of telomeres in telomerase-negative human cancer cells. Here, we demonstrate that ATRX and H3.3 co-localize with the telomeric DNA and associated proteins within the promyelocytic leukemia (PML) bodies in mouse ES cells. The assembly of these telomere-associated PML bodies is most prominent at S phase. RNA interference (RNAi)-mediated knockdown of PML expression induces the disassembly of these nuclear bodies and a telomere dysfunction phenotype in mouse ES cells. Loss of function of PML bodies in mouse ES cells also disrupts binding of ATRX/H3.3 and proper establishment of histone methylation pattern at the telomere. Our study demonstrates that PML bodies act as epigenetic regulators by serving as platforms for the assembly of the telomeric chromatin to ensure a faithful inheritance of epigenetic information at the telomere.


Subject(s)
Cell Nucleus Structures/metabolism , Chromatin/metabolism , Embryonic Stem Cells/metabolism , Telomere/metabolism , Animals , Cell Line, Tumor , Cell Nucleus Structures/chemistry , DNA Helicases/analysis , DNA Repair , Epigenesis, Genetic , Histones/analysis , Humans , Mice , NIH 3T3 Cells , Nuclear Proteins/analysis , Nuclear Proteins/physiology , Phenotype , S Phase , X-linked Nuclear Protein
10.
Nucleic Acids Res ; 39(10): 4300-14, 2011 May.
Article in English | MEDLINE | ID: mdl-21296756

ABSTRACT

Nuclear speckles are known to be the storage sites of mRNA splicing regulators. We report here the identification and characterization of a novel speckle protein, referred to as NSrp70, based on its subcellular localization and apparent molecular weight. This protein was first identified as CCDC55 by the National Institutes of Health Mammalian Gene Collection, although its function has not been assigned. NSrp70 was colocalized and physically interacted with SC35 and ASF/SF2 in speckles. NSrp70 has a putative RNA recognition motif, the RS-like region, and two coiled-coil domains, suggesting a role in RNA processing. Accordingly, using CD44, Tra2ß1 and Fas constructs as splicing reporter minigenes, we found that NSrp70 modulated alternative splice site selection in vivo. The C-terminal 10 amino acids (531-540), including (536)RD(537), were identified as a novel nuclear localization signal, and the region spanning 290-471 amino acids was critical for speckle localization and binding to SC35 and ASF/SF2. The N-terminal region (107-161) was essential for the pre-mRNA splicing activity. Finally, we found that knockout of NSrp70 gene in mice led to a lack of progeny, including fetal embryos. Collectively, we demonstrate that NSrp70 is a novel splicing regulator and essentially required early stage of embryonic development.


Subject(s)
Alternative Splicing , Nuclear Proteins/metabolism , RNA Precursors/metabolism , RNA, Messenger/metabolism , Animals , Cell Line , Cell Nucleus Structures/chemistry , Genes, Lethal , Humans , Mice , Mice, Knockout , Nuclear Proteins/analysis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phenotype , Protein Structure, Tertiary , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors
11.
Nucleic Acids Res ; 39(2): 648-65, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20798176

ABSTRACT

Eukaryotic ribosome biogenesis requires the function of a large number of trans-acting factors which interact transiently with the nascent pre-rRNA and dissociate as the ribosomal subunits proceed to maturation and export to the cytoplasm. Loss-of-function mutations in human trans-acting factors or ribosome components may lead to genetic syndromes. In a previous study, we have shown association between the SBDS (Shwachman-Bodian-Diamond syndrome) and NIP7 proteins and that downregulation of SBDS in HEK293 affects gene expression at the transcriptional and translational levels. In this study, we show that downregulation of NIP7 affects pre-rRNA processing, causing an imbalance of the 40S/60S subunit ratio. We also identified defects at the pre-rRNA processing level with a decrease of the 34S pre-rRNA concentration and an increase of the 26S and 21S pre-rRNA concentrations, indicating that processing at site 2 is particularly slower in NIP7-depleted cells and showing that NIP7 is required for maturation of the 18S rRNA. The NIP7 protein is restricted to the nuclear compartment and co-sediments with complexes with molecular masses in the range of 40S-80S, suggesting an association to nucleolar pre-ribosomal particles. Downregulation of NIP7 affects cell proliferation, consistently with an important role for NIP7 in rRNA biosynthesis in human cells.


Subject(s)
Nuclear Proteins/physiology , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal/metabolism , Cell Line , Cell Nucleus Structures/chemistry , Gene Knockdown Techniques , HEK293 Cells , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Poly A-U/metabolism , Poly U/metabolism , Polyribosomes/chemistry , RNA/chemistry , RNA/metabolism , RNA Precursors/chemistry , RNA, Ribosomal/chemistry
12.
Nucleic Acids Res ; 37(4): 1269-79, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19136466

ABSTRACT

FRAXE is a form of mild to moderate mental retardation due to the silencing of the FMR2 gene. The cellular function of FMR2 protein is presently unknown. By analogy with its homologue AF4, FMR2 was supposed to have a role in transcriptional regulation, but robust evidences supporting this hypothesis are lacking. We observed that FMR2 co-localizes with the splicing factor SC35 in nuclear speckles, the nuclear regions where splicing factors are concentrated, assembled and modified. Similarly to what was reported for splicing factors, blocking splicing or transcription leads to the accumulation of FMR2 in enlarged, rounded speckles. FMR2 is also localized in the nucleolus when splicing is blocked. We show here that FMR2 is able to specifically bind the G-quartet-forming RNA structure with high affinity. Remarkably, in vivo, in the presence of FMR2, the ESE action of the G-quartet situated in mRNA of an alternatively spliced exon of a minigene or of the putative target FMR1 appears reduced. Interestingly, FMR1 is silenced in the fragile X syndrome, another form of mental retardation. All together, our findings strongly suggest that FMR2 is an RNA-binding protein, which might be involved in alternative splicing regulation through an interaction with G-quartet RNA structure.


Subject(s)
Alternative Splicing , G-Quadruplexes , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , RNA/chemistry , Animals , Cell Line , Cell Nucleus Structures/chemistry , Cells, Cultured , Fragile X Mental Retardation Protein/genetics , Humans , Mice , Nuclear Proteins/analysis , Nuclear Proteins/chemistry , Protein Structure, Tertiary , RNA/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/chemistry
13.
Nucleic Acids Res ; 36(2): 423-34, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18039709

ABSTRACT

In heat-shocked human cells, heat shock factor 1 activates transcription of tandem arrays of repetitive Satellite III (SatIII) DNA in pericentromeric heterochromatin. Satellite III RNAs remain associated with sites of transcription in nuclear stress bodies (nSBs). Here we use real-time RT-PCR to study the expression of these genomic regions. Transcription is highly asymmetrical and most of the transcripts contain the G-rich strand of the repeat. A low level of G-rich RNAs is detectable in unstressed cells and a 10(4)-fold induction occurs after heat shock. G-rich RNAs are induced by a wide range of stress treatments including heavy metals, UV-C, oxidative and hyper-osmotic stress. Differences exist among stressing agents both for the kinetics and the extent of induction (>100- to 80.000-fold). In all cases, G-rich transcripts are associated with nSBs. On the contrary, C-rich transcripts are almost undetectable in unstressed cells and modestly increase after stress. Production of SatIII RNAs after hyper-osmotic stress depends on the Tonicity Element Binding Protein indicating that activation of the arrays is triggered by different transcription factors. This is the first example of a non-coding RNA whose transcription is controlled by different transcription factors under different growth conditions.


Subject(s)
DNA, Satellite/chemistry , RNA, Untranslated/biosynthesis , Transcriptional Activation , Animals , CHO Cells , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/ultrastructure , Cricetinae , Cricetulus , DNA-Binding Proteins/metabolism , HeLa Cells , Heat Shock Transcription Factors , Heat-Shock Response , Humans , NFATC Transcription Factors/physiology , Osmotic Pressure , RNA, Untranslated/analysis , RNA, Untranslated/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
14.
Biochim Biophys Acta Mol Cell Res ; 1867(4): 118650, 2020 04.
Article in English | MEDLINE | ID: mdl-31953060

ABSTRACT

Transfection is a powerful tool that enables introducing foreign nucleic acids into living cells in order to study the function of a gene product. Ever since the discovery of transfection many side effects or artifacts caused by transfection reagents have been reported. Here, we show that the transfection reagent, JetPRIME alters the localization of the splicing protein SC35 widely used as a nuclear speckle marker. We demonstrate that transfection of plasmids with JetPRIME leads to enlarged SC35 speckles and SC35 cytoplasmic granules. By contrast, transfection of the same plasmid with Lipofectamine 3000 does not have any effect on SC35 localization. The formation of SC35 cytoplasmic granules by JetPRIME-mediated transfection is independent of exogenous expression by plasmid and although similar in morphology they are distinct from P-bodies and stress granules. This method of transfection affected only SC35 and phosphorylated SR proteins but not the nuclear speckles. The JetPRIME-mediated transfection also showed compromised transcription in cells with enlarged SC35 speckles. Our work indicates that the use of JetPRIME alters SC35 localization and can affect gene expression and alternative splicing. Therefore, caution should be exercised when interpreting results after the use of a transient transfection system, particularly when the subject of the study is the function of a protein in the control of gene expression or mRNA splicing.


Subject(s)
Artifacts , Serine-Arginine Splicing Factors/analysis , Transfection , Cell Line, Tumor , Cell Nucleus Structures/chemistry , Cytoplasmic Granules/chemistry , HeLa Cells , Humans , Indicators and Reagents , RNA Splicing , Transcription, Genetic
15.
J Cell Biol ; 152(5): 1099-106, 2001 Mar 05.
Article in English | MEDLINE | ID: mdl-11238464

ABSTRACT

The transcription coactivator and histone acetyltransferase CAMP response element-binding protein (CBP) has been demonstrated to accumulate in promyelocytic leukemia (PML) bodies. We show that this accumulation is cell type specific. In cells where CBP does not normally accumulate in PML bodies, it can be induced to accumulate in PML bodies through overexpression of either CBP or Pml, but not Sp100. Using fluorescence recovery after photobleaching, we demonstrate that CBP moves rapidly into and out of PML bodies. In contrast, Pml and Sp100 are relatively immobile in the nucleoplasm and within PML nuclear bodies. They possess the characteristics expected of proteins that would play a structural role in the integrity of these subnuclear domains. Our results are consistent with CBP being a dynamic component of PML bodies and that the steady-state level in these structures can be modulated by Pml.


Subject(s)
Antigens, Nuclear , Cell Nucleus Structures/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/drug effects , Fluorescence , Fluorescent Antibody Technique , Humans , Interferons/pharmacology , Leukemia, Promyelocytic, Acute/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Matrix/chemistry , Nuclear Matrix/drug effects , Nuclear Matrix/metabolism , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Transport/drug effects , Recombinant Fusion Proteins/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Tumor Suppressor Proteins
16.
Mol Cell Biol ; 26(13): 4998-5014, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16782886

ABSTRACT

The human transcription elongation factor CA150 contains three N-terminal WW domains and six consecutive FF domains. WW and FF domains, versatile modules that mediate protein-protein interactions, are found in nuclear proteins involved in transcription and splicing. CA150 interacts with the splicing factor SF1 and with the phosphorylated C-terminal repeat domain (CTD) of RNA polymerase II (RNAPII) through its WW and FF domains, respectively. WW and FF domains may, therefore, serve to link transcription and splicing components and play a role in coupling transcription and splicing in vivo. In the study presented here, we investigated the subcellular localization and association of CA150 with factors involved in pre-mRNA transcriptional elongation and splicing. Endogenous CA150 colocalized with nuclear speckles, and this was not affected either by inhibition of cellular transcription or by RNAPII CTD phosphorylation. FF domains are essential for the colocalization to speckles, while WW domains are not required for colocalization. We also performed biochemical assays to understand the role of WW and FF domains in mediating the assembly of transcription and splicing components into higher-order complexes. Transcription and splicing components bound to a region in the amino-terminal part of CA150 that contains the three WW domains; however, we identified a region of the C-terminal FF domains that was also critical. Our results suggest that sequences located at both the amino and carboxyl regions of CA150 are required to assemble transcription/splicing complexes, which may be involved in the coupling of those processes.


Subject(s)
Cell Nucleus Structures/chemistry , DNA-Binding Proteins/metabolism , RNA Splicing , Trans-Activators/analysis , Trans-Activators/chemistry , Transcription Factors/metabolism , Transcription, Genetic , Amino Acid Sequence , Cell Nucleus Structures/metabolism , Cells, Cultured , DNA-Binding Proteins/analysis , Humans , Molecular Sequence Data , Protein Interaction Mapping , Protein Structure, Tertiary/genetics , RNA Polymerase II/metabolism , RNA Splicing Factors , Sequence Deletion , Trans-Activators/metabolism , Transcription Factors/analysis , Transcriptional Elongation Factors
17.
Leuk Res ; 32(10): 1582-92, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18472161

ABSTRACT

Myeloid zinc finger 1 (MZF1) is a transcription factor that plays an important role in blood cell development. Previous reports indicate MZF1 is an essential factor whose abnormal expression results in cancer. However, the molecular mechanisms by which MZF1 functions in development and contributes to cancer progression remain unknown. MZF1 is a member of the SCAN domain family of zinc finger proteins (SCAN-ZFP) that form dimers via their highly conserved SCAN motif. To better understand the molecular mechanism of MZF1 function, we sought to characterize the cellular localization pattern of MZF1 in the context of SCAN dimerization. Here we provide evidence that MZF1 is a constituent of promyelocytic leukemia nuclear bodies (PML-NBs) and that the SCAN domain is necessary for association with these intranuclear structures. In addition, the SCAN-ZFP member ZNF24 was identified as a novel heterodimeric partner of MZF1 that also associates with PML-NBs in a unique ring-type pattern. Finally, we provide support that MZF1 protein may be modified by SUMOylation, which provides further support for localization of MZF1 protein complexes to PML-NBs. Altogether, these data suggest that MZF1 is recruited to PML-NBs and that the SCAN domain may play an integral role in regulating the localization of heterodimeric protein complexes to these intranuclear structures.


Subject(s)
Cell Nucleus Structures/chemistry , Kruppel-Like Transcription Factors/analysis , Kruppel-Like Transcription Factors/chemistry , Cell Line, Tumor , Dimerization , Humans , Kruppel-Like Transcription Factors/metabolism , Nuclear Proteins/analysis , Protein Structure, Tertiary , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/analysis , Zinc Fingers
18.
Biochem J ; 407(3): 407-17, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17650072

ABSTRACT

In vitro, small Hsps (heat-shock proteins) have been shown to have chaperone function capable of keeping unfolded proteins in a form competent for Hsp70-dependent refolding. However, this has never been confirmed in living mammalian cells. In the present study, we show that Hsp27 (HspB1) translocates into the nucleus upon heat shock, where it forms granules that co-localize with IGCs (interchromatin granule clusters). Although heat-induced changes in the oligomerization status of Hsp27 correlate with its phosphorylation and nuclear translocation, Hsp27 phosphorylation alone is not sufficient for effective nuclear translocation of HspB1. Using firefly luciferase as a heat-sensitive reporter protein, we demonstrate that HspB1 expression in HspB1-deficient fibroblasts enhances protein refolding after heat shock. The positive effect of HspB1 on refolding is completely diminished by overexpression of Bag-1 (Bcl-2-associated athanogene), the negative regulator of Hsp70, consistent with the idea of HspB1 being the substrate holder for Hsp70. Although HspB1 and luciferase both accumulate in nuclear granules after heat shock, our results suggest that this is not related to the refolding activity of HspB1. Rather, granular accumulation may reflect a situation of failed refolding where the substrate is stored for subsequent degradation. Consistently, we found 20S proteasomes concentrated in nuclear granules of HspB1 after heat shock. We conclude that HspB1 contributes to an increased chaperone capacity of cells by binding unfolded proteins that are hereby kept competent for refolding by Hsp70 or that are sorted to nuclear granules if such refolding fails.


Subject(s)
Heat-Shock Proteins/physiology , Heat-Shock Response/physiology , Intracellular Fluid/physiology , Molecular Chaperones/physiology , Neoplasm Proteins/physiology , Animals , Cell Line , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/metabolism , HSP27 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/physiology , Intracellular Fluid/chemistry , Mice , Oxidative Stress/physiology , Protein Folding , Protein Sorting Signals/physiology , Protein Transport/physiology , Rats
19.
Nucleic Acids Res ; 34(19): 5508-14, 2006.
Article in English | MEDLINE | ID: mdl-17020918

ABSTRACT

Exposure of cells to stressful conditions elicits a highly conserved defense mechanism termed the heat shock response, resulting in the production of specialized proteins which protect the cells against the deleterious effects of stress. The heat shock response involves not only a widespread inhibition of the ongoing transcription and activation of heat shock genes, but also important changes in post-transcriptional processing. In particular, a blockade in splicing and other post-transcriptional processing has been described following stress in different organisms, together with an altered spatial distribution of the proteins involved in these activities. However, the specific mechanisms that regulate these activities under conditions of stress are little understood. Non-coding RNA molecules are increasingly known to be involved in the regulation of various activities in the cell, ranging from chromatin structure to splicing and RNA degradation. In this review, we consider two non-coding RNAs, the hsr(omega) transcripts in Drosophila and the sat III transcripts in human cells, that seem to be involved in the dynamics of RNA-processing factors in normal and/or stressed cells, and thus provide new paradigms for understanding transcriptional and post-transcriptional regulations in normal and stressed cells.


Subject(s)
Drosophila melanogaster/genetics , Heat-Shock Response , RNA Processing, Post-Transcriptional , RNA, Untranslated/metabolism , Animals , Cell Nucleus Structures/chemistry , DNA, Satellite/genetics , Drosophila melanogaster/chemistry , Drosophila melanogaster/metabolism , Humans , Models, Genetic , RNA, Untranslated/analysis , RNA, Untranslated/physiology
20.
Front Biosci ; 12: 2085-95, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17127446

ABSTRACT

Polyalanine expansion mutations have been identified in eight transcription factors that are associated with a range of congenital disorders. While some of these mutant proteins have been shown to generate cellular aggregates in heterologous cell lines, little is known about the mechanism by which these aggregates cause disease. Here we examine the aggregation and functional properties of the two known polyalanine expansion mutations associated with X-linked Hypopituitarism (XH), SOX3(22Ala) and SOX3(26Ala), which contain an additional seven and eleven alanine residues, respectively. SOX3(22Ala) and SOX3(26Ala) proteins form cytoplasmic aggregates and nuclear inclusions in transiently transfected COS-7 and CHO K1 cells, and in transfected explant cultures of chick neural epithelium. SOX3(26Ala) exhibits a more potent aggregation phenotype, resulting in significantly more cells with dispersed cytoplasmic and large perinuclear aggregates. SOX3(22Ala) and SOX3(26Ala) protein aggregates exhibit the key properties of aggresomes including vimentin redistribution, colocalisation with the Microtubule Organising Centre and sensitivity to microtubule disruption. This is the first time that aggresomes have been implicated in the aetiology of a polyalanine expansion disorder, suggesting that XH and protein conformation disorders may become manifest through similar pathological mechanisms. Further, we show that mutant SOX3 proteins have impaired transcriptional activity and reduced capacity to inhibit beta-catenin/TCF-mediated transcription. These data suggest that deregulation of SOX3 target genes and inappropriate canonical Wnt signaling in central nervous system (CNS) progenitors may also contribute to dysfunction of the hypothalamic-pituitary axis in XH patients.


Subject(s)
DNA Repeat Expansion , DNA-Binding Proteins/genetics , Genetic Diseases, X-Linked/genetics , High Mobility Group Proteins/genetics , Hypopituitarism/genetics , Peptides/genetics , Transcription Factors/genetics , Animals , COS Cells , Cell Nucleus Structures/chemistry , Chick Embryo , Chlorocebus aethiops , Cytoplasmic Structures/chemistry , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/analysis , High Mobility Group Proteins/metabolism , Humans , Male , Peptides/chemistry , SOXB1 Transcription Factors , Transcription Factors/analysis , Transcription Factors/metabolism , Transcriptional Activation , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL