Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
3.
Bone Marrow Transplant ; 52(4): 580-587, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27991894

ABSTRACT

We hypothesized that IV busulfan (Bu) dosing could be safely intensified through pharmacokinetic (PK-) dose guidance to minimize the inter-patient variability in systemic exposure (SE) associated with body-sized dosing, and that this should improve outcome of AML/MDS patients undergoing allogeneic stem cell transplantation. To test this hypothesis, we treated 218 patients (median age 50.7 years, male/female 50/50%) with fludarabine 40 mg/m2 once daily x4, each dose followed by IV Bu, randomized to 130 mg/m2 (N=107) or PK-guided to average daily SE, AUC of 6000 µM min (N=111), stratified for remission status and allo-grafting from HLA-matched donors. Toxicity and GvHD rates in the groups were similar; the risk of relapse or treatment-related mortality remained higher in the fixed-dose group throughout the 80-month observation period. Further, PK-guidance yielded safer disease control, leading to improved overall and PFS, most prominently in MDS patients and in AML patients not in remission at allogeneic stem cell transplantation. We conclude that AML/MDS patients receiving pretransplant conditioning treatment with our 4-day regimen may benefit significantly from PK-guided Bu dosing. This could be considered an alternative to fixed-dose delivery since it provides the benefit of precise dose delivery to a predetermined SE without increasing risk(s) of serious toxicity and/or GvHD.


Subject(s)
Busulfan/administration & dosage , Drug Monitoring/methods , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , Transplantation Conditioning/methods , Transplantation, Homologous/methods , Vidarabine/analogs & derivatives , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Busulfan/pharmacokinetics , Busulfan/toxicity , Female , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/mortality , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Myelodysplastic Syndromes/mortality , Recurrence , Survival Analysis , Transplantation Conditioning/mortality , Transplantation, Homologous/mortality , Treatment Outcome , Vidarabine/administration & dosage
5.
Blood Cancer J ; 5: e357, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26473529

ABSTRACT

Romidepsin is a cyclic molecule that inhibits histone deacetylases. It is Food and Drug Administration-approved for treatment of cutaneous and peripheral T-cell lymphoma, but its precise mechanism of action against malignant T cells is unknown. To better understand the biological effects of romidepsin in these cells, we exposed PEER and SUPT1 T-cell lines, and a primary sample from T-cell lymphoma patient (Patient J) to romidepsin. We then examined the consequences in some key oncogenic signaling pathways. Romidepsin displayed IC50 values of 10.8, 7.9 and 7.0 nm in PEER, SUPT1 and Patient J cells, respectively. Strong inhibition of histone deacetylases and demethylases, increased production of reactive oxygen species and decreased mitochondrial membrane potential were observed, which may contribute to the observed DNA-damage response and apoptosis. The stress-activated protein kinase/c-Jun N-terminal kinase signaling pathway and unfolded protein response in the endoplasmic reticulum were activated, whereas the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) and ß-catenin pro-survival pathways were inhibited. The decreased level of ß-catenin correlated with the upregulation of its inhibitor SFRP1 through romidepsin-mediated hypomethylation of its gene promoter. Our results provide new insights into how romidepsin invokes malignant T-cell killing, show evidence of its associated DNA hypomethylating activity and offer a rationale for the development of romidepsin-containing combination therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Depsipeptides/pharmacology , Lymphoma, T-Cell , Signal Transduction/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Real-Time Polymerase Chain Reaction
6.
Blood Cancer J ; 4: e171, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24413065

ABSTRACT

Treatments for lymphomas include gemcitabine (Gem) and clofarabine (Clo) which inhibit DNA synthesis. To improve their cytotoxicity, we studied their synergism with the alkyl phospholipid edelfosine (Ed). Exposure of the J45.01 and SUP-T1 (T-cell) and the OCI-LY10 (B-cell) lymphoma cell lines to IC10-IC20 levels of the drugs resulted in strong synergistic cytotoxicity for the 3-drug combination based on various assays of cell proliferation and apoptosis. Cell death correlated with increased phosphorylation of histone 2AX and KAP1, decreased mitochondrial transmembrane potential, increased production of reactive oxygen species and release of pro-apoptotic factors. Caspase 8-negative I9.2 cells were considerably more resistant to [Gem+Clo+Ed] than caspase 8-positive cells. In all three cell lines [Gem+Clo+Ed] decreased the level of phosphorylation of the pro-survival protein AKT and activated the stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) stress signaling pathway, which in J45.01 cells resulted in the phosphorylation and heterodimerization of the transcription factors ATF2 and c-Jun. The observed rational mechanism-based efficacy of [Gem+Clo+Ed] based on the synergistic convergence of several pro-death and anti-apoptotic signaling pathways in three very different cell backgrounds provides a powerful foundation for undertaking clinical trials of this drug combination for the treatment of lymphomas.

7.
Biosens Bioelectron ; 41: 862-6, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-22964383

ABSTRACT

The medical diagnostic, the industry, and the biotechnology require rapid, sensitive, and easy to use methods for trypsin activity determination. A simple approach, which meets all these requirements, based on Quartz Crystal Microbalance (QCM) was developed, analytically characterized and described in the present work. QCM application allows rapid trypsin activity evaluation by real time monitoring of the enzymatic degradation of the substrate. The new approach suggested in this work takes advantage of nanoparticles loaded gelatin employment as a trypsin substrate, deposited on the QCM crystal. The heavy nanoparticles leave the substrate layer together with the products of its enzymatic degradation provoking thus a greater decrease of the total QCM crystal mass compared with the non charged substrate. As a result, a higher sensor frequency response occurs. A 10 fold improvement of the LOD was achieved for trypsin activity evaluation applying the proposed method with Ag nanoparticles loaded gelatin (7.5×10(-4) U mL(-1) vs. 7.5×10(-3) U mL(-1) obtained by the "classic" QCM method). The approach subject of this work can be applied with any substrate degrading enzyme.


Subject(s)
Biosensing Techniques/instrumentation , Micro-Electrical-Mechanical Systems/instrumentation , Nanoparticles/chemistry , Nanotechnology/instrumentation , Silver/chemistry , Trypsin/analysis , Trypsin/chemistry , Enzyme Activation , Equipment Design , Equipment Failure Analysis , Nanoparticles/ultrastructure , Reproducibility of Results , Sensitivity and Specificity
8.
Biochem Biophys Res Commun ; 283(2): 430-6, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11327720

ABSTRACT

Nucleolar protein p40/EBP2 is a proliferation-associated antigen that interacts with Epstein-Barr virus nuclear antigen 1 (EBNA1) to maintain the Epstein-Barr virus (EBV) episomes. The yeast p40/EBP2 functions in the processing of 27S-A into 27S-B ribosomal RNA. The present study reports high evolutionary conservation of the cDNA-derived amino acid sequences of p40/EBP2 from frog, chicken, pig, rat, mouse, bovine, and human. p40/EBP2 is ubiquitously expressed in human tissues. It is highly expressed in myelogenous leukemia K-562 compared to other cell lines tested. The human p40/EBP2 gene is located in chromosome 1 with nine exons and eight introns. The minimal promoter region resides 300 nucleotides upstream of a putative ATG initiation codon preceded by a pyrimidine-rich region. These two regions contain eight Sp1 and four c-Ets-1 putative binding sites. Analysis of the p40/EBP2 gene and its promoter region will facilitate studies on the regulation of its expression in EBV-infected and noninfected cells.


Subject(s)
Carrier Proteins/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Nuclear Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/metabolism , Cattle , Cloning, Molecular , DNA, Complementary/genetics , Exons , Gene Expression , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Introns , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , RNA-Binding Proteins , Rats , Saccharomyces cerevisiae Proteins , Sequence Homology, Amino Acid , Tissue Distribution
9.
Virus Res ; 76(1): 17-29, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11376843

ABSTRACT

Previous results from our laboratory have identified a small (60 nt) RNA from the yeast S. cerevisiae that specifically inhibits internal ribosome entry site (IRES)-mediated translation programmed by poliovirus (PV) and hepatitis C virus (HCV) 5'-untranslated region (5'UTR). The yeast inhibitor RNA (called IRNA) was found to efficiently compete with viral 5'UTR for binding of several cellular polypeptides that presumably play important roles in IRES-mediated translation. One such IRNA (and 5'UTR)-binding protein has previously been identified as the La autoantigen. In this report, we have identified a 110-kDa IRNA-binding protein (which also interacts with viral 5'UTR) as nucleolin, a nucleolar RNA binding protein that was previously shown to translocate into the cytoplasm following infection of cells with poliovirus. We demonstrate that nucleolin (called C23) stimulates viral IRES-mediated translation both in vitro and in vivo. We also show that nucleolin mutants containing the carboxy-terminal RNA binding domains but lacking the amino terminal domain inhibit IRES-mediated translation in vitro. The translation inhibitory activity of these mutants correlates with their ability to bind the 5'UTR sequence. These results suggest a role of nucleolin/C23 in viral IRES-mediated translation.


Subject(s)
Gene Expression Regulation, Viral , Phosphoproteins/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Ribosomes/metabolism , 5' Untranslated Regions/genetics , 5' Untranslated Regions/metabolism , Genes, Dominant/genetics , HeLa Cells , Humans , Molecular Weight , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Poliovirus/genetics , Protein Binding , Protein Structure, Tertiary , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae/genetics , Sequence Deletion , Viral Proteins/biosynthesis , Viral Proteins/genetics , Nucleolin
10.
Biochem Biophys Res Commun ; 281(4): 1006-11, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11237763

ABSTRACT

Human RNA helicase II/Gu (hRH II/Gu) protein unwinds double-stranded RNA, folds single-stranded RNA, and may play important roles in ribosomal RNA biogenesis, RNA editing, RNA transport, and general transcription. The genomic organization and the regulation of expression of the hRH II/Gu gene were analyzed. The hRH II/Gu gene is over 29 kb in length, and includes 15 exons. All intron-exon junctions contain consensus GT-AG sequences. An 85 bp basic promoter was identified which lacks a functional TATA box. In LNCaP cells, the hRH II/Gu promoter was up to 50 times as active as pGL2-control vector which has an SV 40 early promoter. Mutation of a putative c-Myc/USF binding site in the region between -110 and -85 caused a decrease in the promoter activity by 2- to 3-fold. A negative regulatory region was also found between -324 and -264. Studies of transcriptional regulation of hRH II/Gu under different physiological and pathophysiological conditions may provide clues to its biological function.


Subject(s)
DNA-Binding Proteins , Genes/genetics , RNA Helicases/genetics , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Exons , HeLa Cells , Humans , Introns , Luciferases/genetics , Luciferases/metabolism , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Sequence Analysis, DNA , Sequence Deletion , Transcription Factors/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Upstream Stimulatory Factors
11.
Mol Immunol ; 37(7): 351-9, 2000 May.
Article in English | MEDLINE | ID: mdl-11074253

ABSTRACT

RNA helicase II/Gu (RH II/Gu) is a nucleolar antigen originally identified using an autoimmune serum from a patient with watermelon stomach. A later report showed that anti-RH II/Gu autoantibodies were also present at low frequency in connective tissue disease (CTD) patients who did not show any symptoms suggestive of a watermelon stomach lesion. In an attempt to understand the relationship between watermelon stomach, also called gastric antral vascular ectasia (GAVE), and autoimmune disorder, we identified the antigenic sites recognized by these autoantibodies. Serum Gu uniquely recognized epitopes at amino acids 646-748 of RH II/Gu and all four CTD patient sera recognized antigenic sites within amino acids 1-173. Anti-RH II/Gu serum produced by immunizing rabbit with recombinant human RH II/Gu protein bound to the same antigenic sites recognized by the CTD patient sera, but it did not recognize the serum Gu epitopes. Results are also presented showing the use of these anti-RH II/Gu antibodies in the analysis of the evolutionary conservation of RH II/Gu in human, monkey and mouse.


Subject(s)
Epitopes, B-Lymphocyte/immunology , Gastric Antral Vascular Ectasia/immunology , Nuclear Proteins/immunology , RNA Helicases/immunology , Amino Acid Sequence , Animals , Autoantibodies/blood , Autoantibodies/immunology , Connective Tissue Diseases/immunology , Conserved Sequence , Epitope Mapping/methods , Epitopes, B-Lymphocyte/genetics , Gastric Antral Vascular Ectasia/blood , Humans , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , RNA Helicases/genetics , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Analysis, Protein/methods , Sequence Homology, Amino Acid
12.
Eur J Biochem ; 267(21): 6395-402, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11029582

ABSTRACT

RNA helicase II/Gu (RH II/Gu) is a nucleolar protein that unwinds dsRNA in a 5' to 3' direction, and introduces a secondary structure into a ssRNA. The helicase domain is at the N-terminal three-quarters of the molecule and the foldase domain is at the C-terminal quarter. The RNA folding activity of RH II/Gu is not a mere artifact of its binding to RNA. This study narrows down the RNA foldase domain to amino acids 749-801 at the C-terminus of the protein. Dissection of this region by deletion and site-directed mutagenesis shows that the four FRGQR repeats, as well as the C-terminal end bind RNA independently. These juxtaposed subdomains are both important for the RNA foldase activity of RH II/Gu. Mutation of either repeat 2 or repeat 4, or simultaneous mutation of Lys792, Arg793 and Lys797 at the C-terminal end of RH II/Gu to alanines inhibits RNA foldase activity. The last 17 amino acids of RH II/Gu can be replaced by an RNA binding motif from nucleolar protein p120 without a deleterious effect on its foldase activity. A model is proposed to explain how RH II/Gu binds and folds an RNA substrate.


Subject(s)
Nucleic Acid Conformation , RNA Helicases/chemistry , RNA Helicases/metabolism , RNA/chemistry , RNA/metabolism , Amino Acid Sequence , Base Sequence , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed/genetics , Protein Binding , Protein Structure, Tertiary , RNA/genetics , RNA Helicases/genetics , Repetitive Sequences, Amino Acid/genetics , Sequence Alignment , Sequence Deletion/genetics , Substrate Specificity
13.
Genomics ; 66(2): 184-94, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10860663

ABSTRACT

RNA helicase II/Gu (RH II/Gu) is a mammalian nucleolar RNA helicase previously identified using an autoimmune serum from a patient with watermelon stomach disease. RH II/Gu can unwind double-stranded RNA and can fold or introduce a secondary structure to a single-stranded RNA. These two enzymatic activities reside in two separate domains of the RH II/Gu molecule. The present study reports the molecular analysis of the cDNA and genomic sequences of the mouse RH II/Gu, its chromosomal localization, and the regulation of expression. The cDNA-derived amino acid sequence shows three tandem repeats at the NH(2)-terminal end of the protein, which are not conserved in the human homologue. Each repeat has 37 amino acids that are rich in basic residues. The helicase and foldase domains are highly conserved between the mouse and the human RH II/Gu. The basic promoter region of the mouse RH II/Gu gene is within 300 nucleotides upstream of a putative ATG initiation codon. Upstream of this promoter region is a silencer that represses transcription of the mouse RH II/Gu gene. This inhibitory region contains three 38-nucleotide repeats in tandem. The mouse RH II/Gu consists of 14 exons and 13 introns. The 3' flanking sequence of the gene contains three putative polyadenylation sites but only two sites are probably functional as shown by Northern blot analysis and 3' end sequences of mouse RH II/Gu cDNA in the EST database. These two alternative polyadenylation sites are approximately 240 and 2100 nucleotides from the TGA stop codon. Both mouse and human RH II/Gu genes are localized on chromosome 10. The availability of the mouse RH II/Gu gene will facilitate its functional analysis including creation of a mouse deficient in RH II/Gu protein.


Subject(s)
Chromosome Mapping/veterinary , Gene Expression Regulation, Enzymologic , RNA Helicases/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , DNA, Complementary , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA Helicases/chemistry , Sequence Homology, Amino Acid , Transcription, Genetic
14.
Biochem Biophys Res Commun ; 266(2): 361-5, 1999 Dec 20.
Article in English | MEDLINE | ID: mdl-10600508

ABSTRACT

RNA helicases are enzymes important in RNA synthesis, processing, transport, and turnover. Human nucleolar RNA helicase II/Gu protein (RH II/Gu) was expressed in a baculovirus system. The purified recombinant RH II/Gu protein has RNA helicase activity on a 5' tailed ds RNA substrate in vitro. We found that Adriamycin, a widely used anticancer drug, inhibited RH II/Gu helicase activity in a dose-dependent manner with an IC(50) of 40 microM. Adriamycin bound to the RNA substrate, and the binding was disrupted by boiling or treatment with 1% SDS, suggesting that the binding of Adriamycin to RNA is reversible. Adriamycin was also found by gel electrophoresis to bind to yeast tRNA to form slow-migrating complexes. These results suggest that Adriamycin can inhibit RNA synthesis or processing by binding to RNA substrates.


Subject(s)
Doxorubicin/pharmacology , RNA Helicases/antagonists & inhibitors , Small Ubiquitin-Related Modifier Proteins , Antineoplastic Agents/pharmacology , Binding, Competitive , Carrier Proteins/metabolism , Humans , Protein Inhibitors of Activated STAT , RNA/metabolism , RNA, Transfer/metabolism , Recombinant Proteins
16.
Exp Cell Res ; 247(2): 389-98, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10066367

ABSTRACT

RNA helicase II/Gu (RH-II/Gu) is a nucleolar RNA helicase of the DEAD-box superfamily. In this study, the functional domains of RH-II/Gu molecule were mapped by fusing the protein or its deletion mutants with a green fluorescence protein and subsequently transfecting or microinjecting the recombinant constructs into HeLa cells. In addition to the identification of a nuclear localization signal (NLS) in the N-terminus and a nucleolar targeting signal in the central helicase domain, a hidden NLS and a nucleolar targeting signal were found in the C-terminal arginine/glycine-rich domain. RH-II/Gu colocalized with fibrillarin, a component of the dense fibrillar region of the nucleolus. Overexpression of the entire RH-II/Gu protein or specific domains of the protein in HeLa cells did not interfere with the normal distribution of fibrillarin. However, when the helicase domain was truncated, the distribution pattern of fibrillarin was distorted. Microinjection of the wild-type RH-II/Gu cDNA into the nucleus of HeLa cells did not disrupt normal cell growth. However, when cells were injected with mutant DNA, only a small percentage of HeLa cells progressed through the cell cycle. Analysis of centrosomes in transfected cells demonstrated that most of the mutant-expressing cells were arrested early in the cell cycle. The results suggest that each of the structural domains of RH-II/Gu is necessary for cell growth and cell cycle progression.


Subject(s)
Chromosome Mapping , Nuclear Proteins/genetics , RNA Helicases/genetics , Binding Sites , Cell Cycle , Cell Division , Cell Nucleolus/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Nuclear Localization Signals , Nuclear Proteins/metabolism , RNA Helicases/metabolism
17.
Biotechniques ; 24(6): 1032-6, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9631199

ABSTRACT

We have constructed a human osteogenic sarcoma cell line, U-2 OS/GFP-Gu, that expresses nucleolar RNA helicase RH-II/Gu tagged with green fluorescent protein (GFP). The presence of a GFP tag does not inhibit RNA helicase, RNA folding and ATPase activities of RH-II/Gu protein. The derived cell line responds to cytotoxic agents like the parental cell line U-2 OS. In the presence of either actinomycin D or toyocamycin, the GFP-RH-II/Gu fusion protein translocates from the nucleolus to the nucleoplasm in the same way as the translocation of endogenous RH-II/Gu. The drug-induced translocation of GFP-RH-II/Gu is easily monitored by direct observation of live cells in vivo. This cell line can be used to screen cytotoxic drugs and to study the mechanisms of drug-induced translocation of RH-II/Gu. The cellular localization of RH-II/Gu during the cell cycle-dependent formation of the nucleolus is readily monitored. Real-time results are obtained more quickly without the disadvantages associated with cell fixation and immunofluorescence-based staining.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleolus/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Nuclear Proteins/genetics , RNA Nucleotidyltransferases/genetics , Small Ubiquitin-Related Modifier Proteins , Biological Transport/drug effects , Biological Transport/genetics , Carrier Proteins/drug effects , Cell Nucleolus/enzymology , Cell Nucleolus/metabolism , Dactinomycin/pharmacology , Green Fluorescent Proteins , Humans , Nuclear Proteins/metabolism , Osteosarcoma , Protein Inhibitors of Activated STAT , RNA Helicases , RNA Nucleotidyltransferases/drug effects , RNA Nucleotidyltransferases/metabolism , Toyocamycin/pharmacology , Tumor Cells, Cultured , Zinc Fingers
18.
Biochem J ; 331 ( Pt 2): 387-93, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9531475

ABSTRACT

Human proliferation-associated protein p120 has previously been shown to localize to the nucleolus, and several functional domains of p120 have been elucidated. By using a nitrocellulose filter binding assay and a Northwestern blotting procedure this study shows that recombinant p120 binds to an rRNA fragment in vitro with a dissociation constant of 4 nM. The specific RNA-binding region of p120 (residues 1-57) was identified with glutathione S-transferase-fused p120 deletion constructs and Northwestern blotting procedures. This RNA-binding region of p120, which includes the nucleolar localization signal of p120, is similar to the arginine-rich RNA-binding regions found in other RNA-binding proteins such as HIV Rev and Tat. Experiments in vivo with HeLa cell nucleolar extracts showed that p120 was associated with the 60-80S pre-ribosomal particles. This association is disrupted by treatment with either RNase A or buffer of high ionic strength. These results suggest that p120 might be involved in rRNA/ribosome maturation, consistent with the role of the yeast homologue Nop2p in rRNA biogenesis.


Subject(s)
Arginine/analysis , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA, Ribosomal/metabolism , Amino Acid Sequence , Binding Sites , Binding, Competitive , Blotting, Northern , Blotting, Western , Cell Nucleolus/chemistry , Gene Deletion , Glutathione Transferase/genetics , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Protein Precursors/metabolism , Recombinant Fusion Proteins , Recombinant Proteins/metabolism , Ribonucleoproteins/metabolism , tRNA Methyltransferases
19.
Exp Cell Res ; 235(2): 413-20, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9299166

ABSTRACT

Some cytotoxic drugs cause translocation of nucleophosmin/B23 and other nucleolar proteins to the nucleoplasm. The present study shows that these drugs caused a similar translocation of RH-II/Gu, a nucleolar RNA helicase. Other nucleolar proteins including p120, UBF, RNA polymerase I large subunit, fibrillarin, p40, and Ren-1 did not translocate. A 2-h treatment of MCF-7 breast cancer cells with 0.008 or 0.16 microM actinomycin D resulted in translocation of RH-II/Gu to the nucleoplasm; these effects were not reversed by 100 microM guanosine. The effects of 0.008 microM actinomycin D, but not 0.16 microM actinomycin D, on the translocation of RH-II/Gu were reversed when the drug was removed. However, the effects of 0.008 or 0.16 microM actinomycin D on the translocation of nucleophosmin/B23 were not reversible. The translocation effects of 50 microM toyocamycin on RH-II/Gu were reversed when the drug was replaced with fresh medium. RH-II/Gu mostly relocalized to the nucleoli within 15 min after toyocamycin was withdrawn; only partial relocalization of nucleophosmin/B23 occurred 40 h after removal of the drug. The effects of toyocamycin were not blocked by 100 microM guanosine. Mycophenolic acid (50 microM, 2-h treatment) caused partial translocation of RH-II/Gu; this effect was slowly reversed upon drug removal and was inhibited by 100 microM guanosine, in a manner similar to the effects of mycophenolic acid on the localization of nucleophosmin/B23. This study shows similarities and differences in the drug-induced translocation and relocalization of RH-II/Gu and nucleophosmin/B23. Analysis of translocation of specific nucleolar proteins may offer a quantitative approach to assessment of potency and duration of effects of cytotoxic agents.


Subject(s)
Adenocarcinoma/enzymology , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/enzymology , RNA Nucleotidyltransferases/metabolism , Biological Transport/drug effects , Cell Nucleolus/enzymology , Dactinomycin/pharmacology , Guanosine/pharmacology , Humans , Mycophenolic Acid/pharmacology , RNA Helicases , Toyocamycin/pharmacology , Tumor Cells, Cultured
20.
Biochem Mol Biol Int ; 42(5): 927-35, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9285060

ABSTRACT

Silver-stainable proteins (SSPs) are aspartic acid-rich nuclear proteins which are silver stained under very specific conditions. Using a degenerate oligodeoxyncleotide probe which codes for acidic amino acid residues, a cDNA for a new SSP, referred to as SSP29, has been isolated. The cDNA-derived amino acid sequence shows SSP29 has a molecular mass of 29 kDa, leucine-rich repeats (LRR) near the NH2-terminal region and acidic clusters at the COOH-terminal portion, indicating that SSP29 is also a member of the LRR subfamily of acidic proteins which have been shown to be involved in antigen-mediated cellular responses, leukemogenesis and differentiation. SSP29 can be stained by Ag-NOR staining. SSP29 is expressed in all human tissues and cell lines tested, localized to nucleoplasm and translocated partially to the nucleoli after heat shock. Its interaction with RNA polymerase I suggests that SSP29 may participate in signal transduction that directs nucleolar activities by regulating ribosomal RNA biosynthesis.


Subject(s)
Neuropeptides , Nuclear Proteins/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA, Complementary/analysis , DNA, Complementary/genetics , HeLa Cells , Humans , Leucine-Rich Repeat Proteins , Molecular Sequence Data , Nuclear Proteins/isolation & purification , Proteins/genetics , Repetitive Sequences, Nucleic Acid , Silver , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL
...