Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Trends Genet ; 34(4): 270-278, 2018 04.
Article in English | MEDLINE | ID: mdl-29329720

ABSTRACT

While the vast majority of cellular DNA in eukaryotes is contained in long linear strands in chromosomes, we have long recognized some exceptions like mitochondrial DNA, plasmids in yeasts, and double minutes (DMs) in cancer cells where the DNA is present in extrachromosomal circles. In addition, specialized extrachromosomal circles of DNA (eccDNA) have been noted to arise from repetitive genomic sequences like telomeric DNA or rDNA. Recently eccDNA arising from unique (nonrepetitive) DNA have been discovered in normal and malignant cells, raising interesting questions about their biogenesis, function and clinical utility. Here, we review recent results and future directions of inquiry on these new forms of eccDNA.


Subject(s)
DNA, Circular/genetics , DNA, Mitochondrial/genetics , DNA, Neoplasm/genetics , Neoplasms/genetics , Neoplastic Cells, Circulating/chemistry , Animals , Chromosomes, Human/chemistry , Chromosomes, Human/metabolism , DNA, Chloroplast/chemistry , DNA, Chloroplast/genetics , DNA, Chloroplast/metabolism , DNA, Circular/chemistry , DNA, Circular/metabolism , DNA, Kinetoplast/chemistry , DNA, Kinetoplast/genetics , DNA, Kinetoplast/metabolism , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/metabolism , DNA, Neoplasm/chemistry , DNA, Neoplasm/metabolism , Eukaryotic Cells/chemistry , Eukaryotic Cells/metabolism , Humans , Kinetoplastida/genetics , Kinetoplastida/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Plants/genetics , Plants/metabolism , Plasmids/chemistry , Plasmids/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/chemistry , Telomere/metabolism
2.
Curr Alzheimer Res ; 13(11): 1267-1276, 2016.
Article in English | MEDLINE | ID: mdl-27357648

ABSTRACT

Several studies suggest that soluble Amyloid ß (Aß) oligomer-induced aberrant neuronal cell cycle re-entry is the initial trigger for a significant part of the neuronal degeneration and loss in Alzheimer's disease (AD). In this study, we investigated the role of Ras, which is a well-known protooncoprotein, in soluble Aß oligomer-induced aberrant neuronal cell cycle activation and subsequent cell loss using retinoic acid differentiated human SH-SY5Y neuroblastoma cells as model system. In line with previous literature, we showed that in vitro preparations of soluble Aß42 oligomers triggered cell cycle activation but not cell proliferation. As a new finding, we showed that Farnesylthiosalicylic acid (FTS), a specific chemical Ras inhibitor, prevented soluble Aß42 oligomer preparation-induced cell cycle activation. Moreover, we showed that the expression of dominant negative mutant H-Ras (S17N) prevented soluble Aß42 oligomer preparation-induced cell cycle activation, confirming the specific role of Ras in this pathway. As a possible better mimic of the situation in the AD brain, we prepared soluble oligomers from Aß42 : Aß40 (3:7) peptide mixture and showed that this oligomer preparation similarly induced cell cycle activation which was also inhibited by the Ras inhibitor. Finally, we showed that FTS prevented soluble Aß42 oligomer preparationinduced cell death in our retinoic acid differentiated SH-SY5Y cells. Overall, our results strongly suggest that Ras activity is required for soluble Aß oligomer-induced aberrant neuronal cell cycle reentry and subsequent neuronal loss, which are considered important mechanisms in AD pathogenesis.


Subject(s)
Amyloid beta-Peptides/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Enzyme Inhibitors/pharmacology , Farnesol/analogs & derivatives , Peptide Fragments/pharmacology , Salicylates/pharmacology , ras Proteins/metabolism , Cell Differentiation , Cell Line, Tumor , Dose-Response Relationship, Drug , Farnesol/pharmacology , Gene Expression Regulation/drug effects , Humans , Microtubule-Associated Proteins/metabolism , Neuroblastoma/pathology , Neurons/drug effects , Time Factors
3.
Cell Cycle ; 15(15): 1986-96, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27254819

ABSTRACT

In eukaryotes, bulk histone expression occurs in the S phase of the cell cycle. This highly conserved system is crucial for genomic stability and proper gene expression. In metazoans, Stem-loop binding protein (SLBP), which binds to 3' ends of canonical histone mRNAs, is a key factor in histone biosynthesis. SLBP is mainly expressed in S phase and this is a major mechanism to limit bulk histone production to the S phase. At the end of S phase, SLBP is rapidly degraded by proteasome, depending on two phosphorylations on Thr 60 and Thr 61. Previously, we showed that SLBP fragment (aa 51-108) fused to GST, is sufficient to mimic the late S phase (S/G2) degradation of SLBP. Here, using this fusion protein as bait, we performed pull-down experiments and found that DCAF11, which is a substrate receptor of CRL4 complexes, binds to the phosphorylated SLBP fragment. We further confirmed the interaction of full-length SLBP with DCAF11 and Cul4A by co-immunoprecipitation experiments. We also showed that DCAF11 cannot bind to the Thr61/Ala mutant SLBP, which is not degraded at the end of S phase. Using ectopic expression and siRNA experiments, we demonstrated that SLBP expression is inversely correlated with DCAF11 levels, consistent with the model that DCAF11 mediates SLBP degradation. Finally, we found that ectopic expression of the S/G2 stable mutant SLBP (Thr61/Ala) is significantly more toxic to the cells, in comparison to wild type SLBP. Overall, we concluded that CRL4-DCAF11 mediates the degradation of SLBP at the end of S phase and this degradation is essential for the viability of cells.


Subject(s)
Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Proteolysis , S Phase , mRNA Cleavage and Polyadenylation Factors/metabolism , Amino Acid Sequence , Cell Death , Cullin Proteins/metabolism , G2 Phase , Gene Knockdown Techniques , HeLa Cells , Humans , Mutant Proteins/metabolism , Nuclear Proteins/chemistry , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA Interference , mRNA Cleavage and Polyadenylation Factors/chemistry
4.
J Cell Biochem ; 115(3): 523-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24122909

ABSTRACT

Histone mRNA levels are cell cycle regulated, and the major regulatory steps are at the posttranscriptional level. A major regulatory mechanism is S-phase restriction of Stem-loop binding protein (SLBP) which binds to the 3' end of histone mRNA and participates in multiple steps of histone mRNA metabolism, including 3' end processing, translation and regulation of mRNA stability. SLBP expression is cell cycle regulated without significant change in its mRNA level. SLBP expression is low in G1 until just before S phase where it functions and at the end of S phase SLBP is degraded by proteasome complex depending on phosphorylations on Thr60 and Thr61. Here using synchronized HeLa cells we showed that SLBP production rate is low in early G1 and recovers back to S phase level somewhere between early and mid-G1. Further, we showed that SLBP is unstable in G1 due to proteasome mediated degradation as a novel mechanism to keep SLBP low in G1. Finally, the S/G2 stable mutant form of SLBP is degraded by proteasome in G1, indicating that indicating that the SLBP degradation in G1 is independent of the previously identified SLBP degradation at S/G2. In conclusion, as a mechanism to limit histone production to S phase, SLBP is kept low in G1 phase due to cooperative action of translation regulation and proteasome mediated degradation which is independent of previously known S/G2 degradation.


Subject(s)
Nuclear Proteins/genetics , Proteasome Endopeptidase Complex/genetics , Protein Biosynthesis , Proteolysis , mRNA Cleavage and Polyadenylation Factors/genetics , G1 Phase/genetics , Gene Expression Regulation , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Nuclear Proteins/metabolism , Phosphorylation/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA, Messenger/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
5.
Cell Cycle ; 9(19): 3857-63, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20935461

ABSTRACT

S phase is characterized by the replication of DNA and assembly of chromatin. This requires the synthesis of large amounts of histone proteins to package the newly replicated DNA. Histone mRNAs are the only mRNAs that do not have polyA tails, ending instead in a conserved stemloop sequence. The stemloop binding protein (SLBP) that binds the 3' end of histone mRNA is cell cycle regulated and SLBP is required in all steps of histone mRNA metabolism. Activation of cyclin E/cdk2 prior to entry into S-phase is critical for initiation of DNA replication and histone mRNA accumulation. At the end of S phase SLBP is rapidly degraded as a result of phosphorylation of SLBP by cyclin A/cdk1 and CK2 effectively shutting off histone mRNA biosynthesis. E2F1, which is required for expression of many S-phase genes, is regulated in parallel with SLBP and its degradation also requires a cyclin binding site, suggesting that it may also be regulated by the same pathway. It is likely that activation of cyclin A/cdk1 helps inhibit both DNA replication and histone mRNA accumulation, marking the end of S phase and entry into G(2)-phase.


Subject(s)
Cyclin A/metabolism , DNA Replication , Histones/genetics , RNA, Messenger/metabolism , S Phase/genetics , Cyclin A/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Gene Expression Regulation , HeLa Cells , Humans
6.
Mol Cell Biol ; 28(14): 4469-79, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18490441

ABSTRACT

Histone mRNA levels are cell cycle regulated, and a major regulatory mechanism is restriction of stem-loop binding protein (SLBP) to S phase. Degradation of SLBP at the end of S phase results in cessation of histone mRNA biosynthesis, preventing accumulation of histone mRNA until SLBP is synthesized just before entry into the next S phase. Degradation of SLBP requires an SFTTP (58 to 62) and KRKL (95 to 98) sequence, which is a putative cyclin binding site. A fusion protein with the 58-amino-acid sequence of SLBP (amino acids 51 to 108) fused to glutathione S-transferase (GST) is sufficient to mimic SLBP degradation at late S phase. Using GST-SLBP fusion proteins as a substrate, we show that cyclin A/Cdk1 phosphorylates Thr61. Furthermore, knockdown of Cdk1 by RNA interference stabilizes SLBP at the end of S phase. Phosphorylation of Thr61 is necessary for subsequent phosphorylation of Thr60 by CK2 in vitro. Inhibitors of CK2 also prevent degradation of SLBP at the end of S phase. Thus, phosphorylation of Thr61 by cyclin A/Cdk1 primes phosphorylation of Thr60 by CK2 and is responsible for initiating SLBP degradation. We conclude that the increase in cyclin A/Cdk1 activity at the end of S phase triggers degradation of SLBP at S/G(2).


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin A/metabolism , Nuclear Proteins/metabolism , S Phase , mRNA Cleavage and Polyadenylation Factors/metabolism , CDC2 Protein Kinase/genetics , Casein Kinase II/metabolism , HeLa Cells , Humans , Phosphorylation , RNA Interference , Threonine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...