Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 20(1): 18, 2022 02 12.
Article in English | MEDLINE | ID: mdl-35151311

ABSTRACT

Resistance to 5-fluorouracil (5-FU) in chemotherapy and recurrence of colorectal tumors is a serious concern that impedes improvements to clinical outcomes. In the present study, we found that conditioned medium (CM) derived from 5-FU-resistant HCT-8/FU cells reduced 5-FU chemosensitivity in HCT-8 colon cancer cells, with corresponding changes to number and morphology of Cajal bodies (CBs) as observable nuclear structures. We found that U2AF homology motif kinase 1 (UHMK1) altered CB disassembly and reassembly and regulated the phosphorylation of coilin, a major component of CBs. This subsequently resulted in a large number of variations in RNA alternative splicing that affected cell survival following 5-FU treatment, induced changes in intracellular phenotype, and transmitted preadaptive signals to adjacent cells in the tumor microenvironment (TME). Our findings suggest that CBs may be useful for indicating drug sensitivity or resistance in tumor cells in response to stress signals. The results also suggest that UHMK1 may be an important factor for maintaining CB structure and morphology by regulating splicing events, especially following cellular exposure to cytotoxic drugs. Video Abstract.


Subject(s)
Coiled Bodies , Colonic Neoplasms , Fluorouracil , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Antimetabolites, Antineoplastic/pharmacology , Coiled Bodies/drug effects , Coiled Bodies/genetics , Coiled Bodies/metabolism , Fluorouracil/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Tumor Microenvironment
2.
Genes Cells ; 26(5): 298-312, 2021 May.
Article in English | MEDLINE | ID: mdl-33608942

ABSTRACT

In eukaryotic nuclei, a number of phase-separated nuclear bodies (NBs) are present. RNA polymerase II (Pol II) is the main player in transcription and forms large condensates in addition to localizing at numerous transcription foci. Cajal bodies (CBs) and histone locus bodies (HLBs) are NBs that are involved in transcriptional and post-transcriptional regulation of small nuclear RNA and histone genes. By live-cell imaging using human HCT116 cells, we here show that Pol II condensates (PCs) nucleated near CBs and HLBs, and the number of PCs increased during S phase concomitantly with the activation period of histone genes. Ternary PC-CB-HLB associates were formed via three pathways: nucleation of PCs and HLBs near CBs, interaction between preformed PC-HLBs with CBs and nucleation of PCs near preformed CB-HLBs. Coilin knockout increased the co-localization rate between PCs and HLBs, whereas the number, nucleation timing and phosphorylation status of PCs remained unchanged. Depletion of PCs did not affect CBs and HLBs. Treatment with 1,6-hexanediol revealed that PCs were more liquid-like than CBs and HLBs. Thus, PCs are dynamic structures often nucleated following the activation of gene clusters associated with other NBs.


Subject(s)
Coiled Bodies/metabolism , Histones/metabolism , RNA Polymerase II/metabolism , Cell Survival/drug effects , Coiled Bodies/drug effects , Glycols/pharmacology , Green Fluorescent Proteins/metabolism , HCT116 Cells , Humans , Models, Biological , Nuclear Proteins/metabolism , S Phase/drug effects
3.
Sci Rep ; 10(1): 10738, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32612161

ABSTRACT

Spinal muscular atrophy (SMA) is a devastating autosomal recessive neuromuscular disease characterized by degeneration of spinal cord alpha motor neurons (αMNs). SMA is caused by the homozygous deletion or mutation of the survival motor neuron 1 (SMN1) gene, resulting in reduced expression of SMN protein, which leads to αMN degeneration and muscle atrophy. The majority of transcripts of a second gene (SMN2) generate an alternative spliced isoform that lacks exon 7 and produces a truncated nonfunctional form of SMN. A major function of SMN is the biogenesis of spliceosomal snRNPs, which are essential components of the pre-mRNA splicing machinery, the spliceosome. In recent years, new potential therapies have been developed to increase SMN levels, including treatment with antisense oligonucleotides (ASOs). The ASO-nusinersen (Spinraza) promotes the inclusion of exon 7 in SMN2 transcripts and notably enhances the production of full-length SMN in mouse models of SMA. In this work, we used the intracerebroventricular injection of nusinersen in the SMN∆7 mouse model of SMA to evaluate the effects of this ASO on the behavior of Cajal bodies (CBs), nuclear structures involved in spliceosomal snRNP biogenesis, and the cellular distribution of polyadenylated mRNAs in αMNs. The administration of nusinersen at postnatal day (P) 1 normalized SMN expression in the spinal cord but not in skeletal muscle, rescued the growth curve and improved motor behavior at P12 (late symptomatic stage). Importantly, this ASO recovered the number of canonical CBs in MNs, significantly reduced the abnormal accumulation of polyadenylated RNAs in nuclear granules, and normalized the expression of the pre-mRNAs encoding chondrolectin and choline acetyltransferase, two key factors for αMN homeostasis. We propose that the splicing modulatory function of nusinersen in SMA αMN is mediated by the rescue of CB biogenesis, resulting in enhanced polyadenylated pre-mRNA transcription and splicing and nuclear export of mature mRNAs for translation. Our results support that the selective restoration of SMN expression in the spinal cord has a beneficial impact not only on αMNs but also on skeletal myofibers. However, the rescue of SMN expression in muscle appears to be necessary for the complete recovery of motor function.


Subject(s)
Coiled Bodies/drug effects , Disease Models, Animal , Motor Neurons/drug effects , Muscular Atrophy, Spinal/prevention & control , Oligonucleotides/pharmacology , RNA, Messenger/drug effects , Survival of Motor Neuron 2 Protein/physiology , Active Transport, Cell Nucleus , Animals , Coiled Bodies/pathology , Mice , Mice, Knockout , Motor Neurons/pathology , Muscular Atrophy, Spinal/etiology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , RNA, Messenger/genetics
4.
Sci Rep ; 8(1): 2075, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391529

ABSTRACT

The hereditary neurodegenerative disorder spinal muscular atrophy (SMA) is characterized by the loss of spinal cord motor neurons and skeletal muscle atrophy. SMA is caused by mutations of the survival motor neuron (SMN) gene leading to a decrease in SMN protein levels. The SMN deficiency alters nuclear body formation and whether it can contribute to the disease remains unclear. Here we screen a series of small-molecules on SMA patient fibroblasts and identify flunarizine that accumulates SMN into Cajal bodies, the nuclear bodies important for the spliceosomal small nuclear RNA (snRNA)-ribonucleoprotein biogenesis. Using histochemistry, real-time RT-PCR and behavioural analyses in a mouse model of SMA, we show that along with the accumulation of SMN into Cajal bodies of spinal cord motor neurons, flunarizine treatment modulates the relative abundance of specific spliceosomal snRNAs in a tissue-dependent manner and can improve the synaptic connections and survival of spinal cord motor neurons. The treatment also protects skeletal muscles from cell death and atrophy, raises the neuromuscular junction maturation and prolongs life span by as much as 40 percent (p < 0.001). Our findings provide a functional link between flunarizine and SMA pathology, highlighting the potential benefits of flunarizine in a novel therapeutic perspective against neurodegenerative diseases.


Subject(s)
Coiled Bodies/drug effects , Flunarizine/pharmacology , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Animals , Cell Line , Coiled Bodies/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Flunarizine/therapeutic use , HeLa Cells , Humans , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/drug therapy , Small Molecule Libraries/pharmacology
5.
Chromosoma ; 121(6): 629-42, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23064547

ABSTRACT

Cajal bodies (CB) are subnuclear domains that contain various proteins with diverse functions including the CB marker protein coilin. In this study, we investigate the proteolytic activity of calpain on coilin. Here, we report a 28-kDa cleaved coilin fragment detected by two coilin antibodies that is cell cycle regulated, with levels that are consistently reduced during mitosis. We further show that an in vitro calpain assay with full-length or C-terminal coilin recombinant protein releases the same size cleaved fragment. Furthermore, addition of exogenous RNA to purified coilin induces proteolysis by calpain. We also report that the relative levels of this cleaved coilin fragment are susceptible to changes induced by various cell stressors, and that coilin localization is affected by inhibition or knockdown of calpain both under normal and stressed conditions. Collectively, our data suggest that coilin is subjected to regulated specific proteolysis by calpain, and this processing may play a role in the regulation of coilin activity and CB formation.


Subject(s)
Coiled Bodies/metabolism , Nuclear Proteins/metabolism , Calpain/antagonists & inhibitors , Calpain/metabolism , Cell Cycle/physiology , Cell Nucleolus/metabolism , Coiled Bodies/drug effects , HeLa Cells/drug effects , Humans , Leupeptins/pharmacology , Nuclear Proteins/genetics , Proteolysis
6.
FEBS Lett ; 586(19): 3404-9, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22986342

ABSTRACT

Coilin is considered the Cajal body (CB) marker protein. In this report, we investigated the role of coilin in the DNA damage response and found that coilin reduction correlated with significantly increased levels of soluble γH2AX in etoposide treated U2OS cells. Additionally, coilin levels influenced the proliferation rate and cell cycle distribution of cells exposed to etoposide. Moreover, coilin overexpression inhibited nucleolar localization of endogenous coilin in etoposide treated U2OS cells. Collectively, these data provide additional evidence for coilin and CBs in the DNA damage response.


Subject(s)
Cell Cycle Checkpoints/physiology , Etoposide/pharmacology , Histones/metabolism , Nuclear Proteins/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Cell Proliferation/drug effects , Coiled Bodies/drug effects , Coiled Bodies/metabolism , DNA Damage , Gene Knockdown Techniques , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA, Small Interfering/genetics , Solubility
7.
J Cell Biochem ; 113(11): 3313-29, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22644811

ABSTRACT

Although it is well known that chromosomes are non-randomly organized during interphase, it is not completely clear whether higher-order chromatin structure is transmitted from mother to daughter cells. Therefore, we addressed the question of how chromatin is rearranged during interphase and whether heterochromatin pattern is transmitted after mitosis. We additionally tested the similarity of chromatin arrangement in sister interphase nuclei. We noticed a very active cell rotation during interphase, especially when histone hyperacetylation was induced or transcription was inhibited. This natural phenomenon can influence the analysis of nuclear arrangement. Using photoconversion of Dendra2-tagged core histone H4 we showed that the distribution of chromatin in daughter interphase nuclei differed from that in mother cells. Similarly, the nuclear distribution of heterochromatin protein 1ß (HP1ß) was not completely identical in mother and daughter cells. However, identity between mother and daughter cells was in many cases evidenced by nucleolar composition. Moreover, morphology of nucleoli, HP1ß protein, Cajal bodies, chromosome territories, and gene transcripts were identical in sister cell nuclei. We conclude that the arrangement of interphase chromatin is not transmitted through mitosis, but the nuclear pattern is identical in naturally synchronized sister cells. It is also necessary to take into account the possibility that cell rotation and the degree of chromatin condensation during functionally specific cell cycle phases might influence our view of nuclear architecture.


Subject(s)
Cell Nucleolus/ultrastructure , Coiled Bodies/ultrastructure , Heterochromatin/genetics , Interphase/genetics , Mitosis/genetics , Animals , Cell Line , Cell Nucleolus/drug effects , Cell Nucleolus/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Coiled Bodies/drug effects , Coiled Bodies/genetics , Dactinomycin/pharmacology , Fluorescent Dyes , Heterochromatin/drug effects , Heterochromatin/ultrastructure , Histone Deacetylase Inhibitors/pharmacology , Histones/genetics , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Interphase/drug effects , Mice , Microscopy, Fluorescence , Mitosis/drug effects , Photochemical Processes , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/biosynthesis
8.
Int J Mol Med ; 29(6): 1031-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22426501

ABSTRACT

Cyclophilin B (CypB) is an ubiquitously expressed protein, which performs several intra- and extracellular functions. Despite its abundant use as a household protein, little is known about its exact cellular localization and dynamics. In the present study we show that endogenous CypB localizes in one of two distinct compartments, either within the endoplasmic reticulum (ER) or inside the nucleus, accumulating in the fibrillar centers of the nucleoli. By means of a genetic deletion screen, we identified a minimal nucleolar localization signal for efficient relocation to the nucleoli. Within the fibrillar centers, CypB colocalized with RNA polymerase, upstream binding factor-1 (UBF), fibrillarin and dyskerin (DCK1). Even after chemical disruption of the nucleoli, a strong interaction with these proteins remained. Using live cell imaging, we showed a persistent colocalization of CypB with proteins involved in the ribosome biogenesis during the transcriptionally more active phases of the cell cycle. Supported by in silico data, our observations suggest that CypB interacts with these proteins and is involved in ribosome biogenesis and RNA transcription.


Subject(s)
Cell Nucleus/genetics , Cyclophilins/metabolism , RNA/genetics , Transcription, Genetic , Cell Nucleolus/metabolism , Cell Nucleus/drug effects , Coiled Bodies/drug effects , Coiled Bodies/metabolism , Computational Biology , Cyclosporine/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fluorescent Antibody Technique , Gene Deletion , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Male , Protein Binding/drug effects , Protein Transport/drug effects , RNA/metabolism , RNA Processing, Post-Transcriptional/drug effects , Recombinant Fusion Proteins/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Time Factors , Transcription, Genetic/drug effects , Transfection
9.
PLoS One ; 6(10): e25743, 2011.
Article in English | MEDLINE | ID: mdl-21991343

ABSTRACT

Coilin is a nuclear phosphoprotein that accumulates in Cajal bodies (CBs). CBs participate in ribonucleoprotein and telomerase biogenesis, and are often found in cells with high transcriptional demands such as neuronal and cancer cells, but can also be observed less frequently in other cell types such as fibroblasts. Many proteins enriched within the CB are phosphorylated, but it is not clear what role this modification has on the activity of these proteins in the CB. Coilin is considered to be the CB marker protein and is essential for proper CB formation and composition in mammalian cells. In order to characterize the role of coilin phosphorylation on CB formation, we evaluated various coilin phosphomutants using transient expression. Additionally, we generated inducible coilin phosphomutant cell lines that, when used in combination with endogenous coilin knockdown, allow for the expression of the phosphomutants at physiological levels. Transient expression of all coilin phosphomutants except the phosphonull mutant (OFF) significantly reduces proliferation. Interestingly, a stable cell line induced to express the coilin S489D phosphomutant displays nucleolar accumulation of the mutant and generates a N-terminal degradation product; neither of which is observed upon transient expression. A N-terminal degradation product and nucleolar localization are also observed in a stable cell line induced to express a coilin phosphonull mutant (OFF). The nucleolar localization of the S489D and OFF coilin mutants observed in the stable cell lines is decreased when endogenous coilin is reduced. Furthermore, all the phosphomutant cells lines show a significant reduction in CB formation when compared to wild-type after endogenous coilin knockdown. Cell proliferation studies on these lines reveal that only wild-type coilin and the OFF mutant are sufficient to rescue the reduction in proliferation associated with endogenous coilin depletion. These results emphasize the role of coilin phosphorylation in the formation and activity of CBs.


Subject(s)
Coiled Bodies/metabolism , Mutant Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Proteolysis , Amino Acid Sequence , Cell Line , Cell Proliferation/drug effects , Coiled Bodies/drug effects , Doxycycline/pharmacology , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Mutant Proteins/chemistry , Nuclear Proteins/chemistry , Phosphoproteins/chemistry , Phosphorylation/drug effects , Proteolysis/drug effects , Recombinant Fusion Proteins/metabolism , Transfection
10.
Biol Pharm Bull ; 30(11): 2037-42, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17978472

ABSTRACT

Although ginsenosides have a variety of physiologic or pharmacologic functions in various regions, there are only a few reports on the effects of ginsenosides on gastrointestinal (GI) motility. We studied the modulation of pacemaker activities by ginseng total saponins in the interstitial cells of Cajal (ICC) using the whole cell patch-clamp technique. Externally applied ginseng total saponins (GTS) produced membrane depolarization in the current-clamp mode and increased tonic inward pacemaker currents in the voltage-clamp mode. The application of flufenamic acid or niflumic acid abolished the generation of pacemaker currents, but only treatment with flufenamic acid inhibited the GTS-induced tonic inward currents. The tonic inward currents induced by GTS were not inhibited by the intracellular application of guanosine 5'-[beta-thio]diphosphate trilithium salt. Pretreatment with a Ca(2+)-free solution, with U-73122, an active phospholipase C inhibitor, and with thapsigargin, a Ca(2')-ATPase inhibitor of the endoplasmic reticulum, abolished the generation of pacemaker currents and suppressed the GTS-induced action. However, treatment with chelerythrine and calphostin C, protein kinase C inhibitors, did not block the GTS-induced effects on the pacemaker currents. These results suggest that ginsenosides modulate the pacemaker activities of the ICC, and the ICC can be targets for ginsenosides, and their interaction can affect intestinal motility.


Subject(s)
Coiled Bodies/drug effects , Intestine, Small/drug effects , Intestine, Small/physiology , Membrane Potentials/drug effects , Panax/chemistry , Saponins/pharmacology , Action Potentials/drug effects , Animals , Electrophysiology , Female , Intestine, Small/cytology , Male , Mice , Mice, Inbred BALB C , Patch-Clamp Techniques
11.
J Cell Biochem ; 98(5): 1320-33, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16514659

ABSTRACT

We have previously shown that ISG20, an interferon (IFN)-induced gene, encodes a 3' to 5' exoribonuclease member of the DEDD superfamily of exonucleases. ISG20 specifically degrades single-stranded RNA. In this report, using immunofluorescence analysis, we demonstrate that in addition to a diffuse cytoplasmic and nucleoplasmic localization, the endogenous ISG20 protein was present in the nucleus both in the nucleolus and in the Cajal bodies (CBs). In addition, we show that the ectopic expression of the CBs signature protein, coilin, fused to the red fluorescent protein (coilin-dsRed) increased the number of nuclear dots containing both ISG20 and coilin-dsRed. Using electron microcopy analysis, ISG20 appeared principally concentrated in the dense fibrillar component of the nucleolus, the major site for rRNA processing. We also present evidences that ISG20 was associated with survival of motor neuron (SMN)-containing macromolecular nuclear complexes required for the biogenesis of various small nuclear ribonucleoproteins. Finally, we demonstrate that ISG20 was associated with U1 and U2 snRNAs, and U3 snoRNA. The accumulation of ISG20 in the CBs after IFN treatment strongly suggests its involvement in a new route for IFN-mediated inhibition of protein synthesis by modulating snRNA and rRNA maturation.


Subject(s)
Cell Nucleolus/enzymology , Coiled Bodies/enzymology , Cyclic AMP Response Element-Binding Protein/metabolism , Exonucleases/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Cell Nucleolus/drug effects , Cell Nucleolus/ultrastructure , Coiled Bodies/drug effects , Coiled Bodies/ultrastructure , Exoribonucleases , HeLa Cells , Humans , Interferons/pharmacology , Microscopy, Immunoelectron , Protein Binding , RNA, Small Nuclear/metabolism , SMN Complex Proteins
12.
J Pharmacol Exp Ther ; 317(1): 439-45, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16330493

ABSTRACT

Substance P is generally considered an excitatory neurotransmitter related to gut motor activity, although an inhibitory influence of neurokinin-1 (NK1) receptor activation on peristalsis has also been reported. With an optimized in vitro method to assess distention-induced peristalsis, our aim was to clarify the effect of NK1 receptor activation on peristaltic activity and to reveal the mechanisms by which NK1 activation alters peristalsis. Distention of the small intestine of the mouse and guinea pig induced periodic occurrence of rhythmic waves of propagating rings of circular muscle contraction, associated with slow waves and superimposed action potentials, that propelled intestinal contents aborally. Activation of NK1 receptors by Ava[l-Pro(9),N-MeLeu10] substance P(7-11) (GR 73632) and Sar(9), Met(O(2))(11) on smooth muscle cells resulted in prolongation of the activity periods and increased action potential generation occurring superimposed on the intestinal slow wave activity. Activation of NK1 receptors on interstitial cells of Cajal resulted in an increase in slow wave frequency. Slow wave amplitude increased, likely by increased cell-to-cell coupling. The NK1 antagonist (S)-1-(2-[3-(3,4-dichlorophenyl)-1-(3-isopropoxyphenylacetyl)piperidin-3-yl]ethyl)-4-phenyl-1-azoniabicyclo[2.2.2]octane chloride (SR 140333) induced a decrease in the slow wave frequency and duration of the activity periods evoked by distention, which makes it likely that NK1 receptor activation plays a role in the normal physiological distention-induced generation of peristaltic motor patterns. In summary, NK1 receptors play a role in normal development of peristalsis and NK1 receptor activation markedly increases propulsive peristaltic contractile activity.


Subject(s)
Coiled Bodies/drug effects , Intestine, Small/drug effects , Muscle, Smooth/drug effects , Peptide Fragments/pharmacology , Peristalsis/drug effects , Receptors, Neurokinin-1/agonists , Substance P/analogs & derivatives , Action Potentials/drug effects , Animals , Coiled Bodies/metabolism , Coiled Bodies/physiology , Dilatation , Female , Guinea Pigs , In Vitro Techniques , Intestine, Small/innervation , Intestine, Small/metabolism , Mice , Muscle, Smooth/innervation , Muscle, Smooth/metabolism , Neurokinin-1 Receptor Antagonists , Piperidines/pharmacology , Quinuclidines/pharmacology , Receptors, Neurokinin-1/physiology , Substance P/pharmacology
13.
Glia ; 40(3): 378-88, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12420317

ABSTRACT

It is well established that forskolin-induced elevation of cAMP results in activation of DNA synthesis in Schwann cell cultures. This promitotic response is partially mediated by the Cdk2, which is required for the transition from the G1 to the S phase of the cell cycle. In the present study, we analyze the effects of cAMP elevation in cultured Schwann cells on the transcriptional activity and on the organization of two nuclear compartments involved in pre-mRNA processing: Cajal bodies (CBs) and splicing factor compartments. Our immunofluorescence and quantitative studies show that forskolin treatment induces a 5.6-fold increase in the proportion of S phase Schwann cells, detected by a short pulse (20 min) of BrdU incorporation. This increase in DNA synthesis correlates with an activation of global transcription, as is indicated by the higher nuclear incorporation of BrU in nascent RNA. Forskolin treatment significantly increases the percentage of Schwann cells containing typical CBs, which concentrate spliceosomal snRNPs and the survival motor neuron (SMN) protein. This increase in the number of CBs closely correlates with the activation of transcription. Moreover, the occurrence of CBs is significantly higher in BrdU (+) cells than in BrdU (-) cells, indicating that entry in the S phase promotes the formation of CBs. During the S phase, Schwann cell nuclei display higher Cdk2 nuclear staining and concentrate this kinase in CBs. Forskolin also induces a redistribution of the pre-mRNA splicing factors in Schwann cells. Primary cultures of Schwann cells provide an excellent physiological model to demonstrate that the assembly of CBs is a transcription- and replication-dependent cellular event. Moreover, the S phase accumulation of Cdk2 observed in Schwann cells supports a functional link between CBs and DNA replication, which is mediated by the possible participation of CBs in the regulatory control of histone gene expression.


Subject(s)
CDC2-CDC28 Kinases , Cell Division/genetics , Coiled Bodies/genetics , Cyclic AMP/metabolism , DNA Replication/genetics , RNA Splicing/genetics , Schwann Cells/metabolism , Animals , Animals, Newborn , Cell Division/drug effects , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cells, Cultured , Coiled Bodies/drug effects , Coiled Bodies/metabolism , Colforsin/pharmacology , Cyclic AMP/pharmacology , Cyclic AMP Response Element-Binding Protein , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/drug effects , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , DNA Replication/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Histones/drug effects , Histones/genetics , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Peripheral Nerves/cytology , Peripheral Nerves/growth & development , Peripheral Nerves/metabolism , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Splicing/drug effects , RNA-Binding Proteins , Rats , Rats, Sprague-Dawley , Ribonucleoproteins, Small Nuclear/drug effects , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , S Phase/drug effects , S Phase/genetics , SMN Complex Proteins , Schwann Cells/cytology , Schwann Cells/drug effects , Survival of Motor Neuron 1 Protein , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
14.
Exp Cell Res ; 279(1): 111-7, 2002 Sep 10.
Article in English | MEDLINE | ID: mdl-12213219

ABSTRACT

Ki67 is a nuclear protein expressed in proliferating cells, but not in quiescent or G(0)-arrested cells. Similar to the proliferating cell nuclear antigen and several other well-characterized molecules, Ki67 exhibits a repeating pattern of regulated expression and redistribution during the cell cycle, making it a useful marker for cell cycle phase. In addition to other structures labeled, concentrated foci may be observed in the nucleus and sometimes the cytoplasm. We observed that these Ki67 foci can be found at any stage of the endothelial cell cycle. They are not coincident with coiled bodies (CB), as determined in double-label immunofluorescence experiments with anti-Ki67 and antibodies to the CB marker protein pigpen. However, arrest of BPA47 endothelial cells in G(1) with amiloride + bumetanide induces colocalization of pigpen and Ki67 in 40% of cells exhibiting Ki67 foci. We conducted a series of experiments to examine the possibilities that pigpen was exported from CB and into unique, Ki67-containing foci or that Ki67 was imported into pigpen-containing CB. Our results showed us that although CB typically contain both coilin and pigpen, amiloride + bumetanide-induced G(1) arrest reconfigured the CB compartment into three populations of foci: one containing pigpen without coilin, the second containing coilin without pigpen, and a third containing both pigpen and coilin together. Furthermore, G(1) arrest resulted in Ki67 redistribution into both coilin- and pigpen-containing foci. The results suggest that under certain conditions, "resident" CB proteins can be differentially redistributed, and proteins not previously recognized as resident in CB can be driven into that compartment. Our observations underscore the fluid nature of CB. They demonstrate that previously reported heterogeneity in the CB compartment can be amplified by a specific experimental manipulation. This may be useful in future analyses of protein trafficking within the CB compartment and between CB and other cellular compartments. Finally, the redistribution of Ki67 into CB represents a new finding for a widely expressed but poorly understood molecule, one that may be useful in elucidating function.


Subject(s)
Amiloride/pharmacology , Bumetanide/pharmacology , Coiled Bodies/chemistry , Animals , Cattle , Cell Cycle , Cell Nucleus/chemistry , Cells, Cultured , Coiled Bodies/drug effects , Coiled Bodies/metabolism , DNA-Binding Proteins , Endothelium, Vascular/chemistry , G1 Phase/drug effects , Ki-67 Antigen/analysis , Ki-67 Antigen/metabolism , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Protein Transport/drug effects , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism
15.
J Cell Sci ; 114(Pt 24): 4407-19, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11792806

ABSTRACT

Splicing snRNPs (small nuclear ribonucleoproteins) are essential sub-units of the spliceosome. Here we report the establishment of stable cell lines expressing fluorescently tagged SmB, a core snRNP protein. Analysis of these stable cell lines has allowed us to characterize the nuclear pathway that leads to snRNP accumulation in nuclear speckles and has identified a limiting nucleolar step in the pathway that can be saturated by overexpression of Sm proteins. After nuclear import, newly assembled snRNPs accumulate first in a subset of Cajal bodies that contain both p80-coilin and the survival of motor neurons protein (SMN) and not in bodies that contain p80-coilin but lack SMN. Treatment of cells with leptomycin B (LMB) inhibits both the accumulation of snRNPs in nuclear bodies and their subsequent accumulation in speckles. The formation of Cajal bodies is enhanced by Sm protein expression and the assembly of new snRNPs. Formation of heterokaryons between HeLa cell lines expressing Sm proteins and primary cells that usually lack Cajal bodies results in the detection of Cajal bodies in primary cell nuclei. Transient over-expression of exogenous SmB alone is sufficient to induce correspondingly transient Cajal body formation in primary cells. These data indicate that the level of snRNP protein expression and snRNP assembly, rather than the expression levels of p80-coilin or SMN, may be a key trigger for Cajal body formation.


Subject(s)
Coiled Bodies/physiology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Ribonucleoproteins, Small Nuclear/biosynthesis , Autoantigens/biosynthesis , Autoantigens/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Cell Fusion , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Coiled Bodies/drug effects , Coiled Bodies/metabolism , Cyclic AMP Response Element-Binding Protein , Fatty Acids, Unsaturated/pharmacology , HeLa Cells , Humans , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Luminescent Proteins/physiology , Muscular Atrophy, Spinal/metabolism , Protein Transport/drug effects , RNA-Binding Proteins , Recombinant Fusion Proteins/biosynthesis , Ribonucleoproteins, Small Nuclear/antagonists & inhibitors , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/physiology , SMN Complex Proteins , Transfection , snRNP Core Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...