Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nat Commun ; 10(1): 2278, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31123254

ABSTRACT

Mammalian spermatogenesis is sustained by mitotic germ cells with self-renewal potential known as undifferentiated spermatogonia. Maintenance of undifferentiated spermatogonia and spermatogenesis is dependent on tightly co-ordinated transcriptional and post-transcriptional mechanisms. The RNA helicase DDX5 is expressed by spermatogonia but roles in spermatogenesis are unexplored. Using an inducible knockout mouse model, we characterise an essential role for DDX5 in spermatogonial maintenance and show that Ddx5 is indispensable for male fertility. We demonstrate that DDX5 regulates appropriate splicing of key genes necessary for spermatogenesis. Moreover, DDX5 regulates expression of cell cycle genes in undifferentiated spermatogonia post-transcriptionally and is required for cell proliferation and survival. DDX5 can also act as a transcriptional co-activator and we demonstrate that DDX5 interacts with PLZF, a transcription factor required for germline maintenance, to co-regulate select target genes. Combined, our data reveal a critical multifunctional role for DDX5 in regulating gene expression programmes and activity of undifferentiated spermatogonia.


Subject(s)
DEAD-box RNA Helicases/metabolism , Promyelocytic Leukemia Zinc Finger Protein/metabolism , RNA Splicing/physiology , Spermatogenesis/genetics , Spermatogonia/metabolism , Animals , Cell Cycle/genetics , Cell Proliferation/genetics , Coculture Techniques , DEAD-box RNA Helicases/genetics , Embryo, Mammalian , Fertility/genetics , Fibroblasts , Gene Expression Regulation/physiology , Male , Mice , Mice, Knockout , Models, Animal , Primary Cell Culture , Testis/cytology
2.
Nature ; 562(7725): 150, 2018 10.
Article in English | MEDLINE | ID: mdl-29973715

ABSTRACT

Change History: This Article has been retracted; see accompanying Retraction. Corrected online 20 January: In this Article, author Frank Rigo was incorrectly listed with a middle initial; this has been corrected in the online versions of the paper.

3.
Ultraschall Med ; 39(4): 422-431, 2018 Aug.
Article in English | MEDLINE | ID: mdl-28934812

ABSTRACT

BACKGROUND: Prediction of pathological complete response (pCR) of primary breast cancer to neoadjuvant chemotherapy (NACT) may influence planned surgical approaches in the breast and axilla. The aim of this project is to assess the value of interim shear wave elastography (SWE), ultrasound (US) and magnetic resonance imaging (MRI) after 3 cycles in predicting pCR. METHODS: 64 patients receiving NACT had baseline and interim US, SWE and MRI examinations. The mean lesion stiffness at SWE, US and MRI diameter was measured at both time points. We compared four parameters with pCR status: a) Interim mean stiffness ≤ or > 50 kPa; b) Percentage stiffness reduction; c) Percentage US diameter reduction and d) Interim MRI response using RECIST criteria. The Chi square test was used to assess significance. RESULTS: Interim stiffness of ≤ or > 50 kPa gave the best prediction of pCR with pCR seen in 10 of 14 (71 %) cancers with an interim stiffness of ≤ 50 kPa, compared to 7 of 50 (14 %) of cancers with an interim stiffness of > 50 kPa, (p < 0.0001) (sensitivity 59 %, specificity 91 %, PPV 71 %, NPV 86 % and diagnostic accuracy 83 %). Percentage reduction in stiffness was the next best parameter (sensitivity 59 %, specificity 85 %, p < 0.0004) followed by reduction in MRI diameter of > 30 % (sensitivity 50 % and specificity 79 %, p = 0.03) and % reduction in US diameter (sensitivity 47 %, specificity 81 %, p = 0.03). Similar results were obtained from ROC analysis. CONCLUSION: SWE stiffness of breast cancers after 3 cycles of NACT and changes in stiffness from baseline are strongly associated with pCR after 6 cycles.


Subject(s)
Breast Neoplasms , Elasticity Imaging Techniques , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Female , Humans , Magnetic Resonance Imaging , Neoadjuvant Therapy , Ultrasonography
4.
Nucleic Acids Res ; 45(19): 11056-11069, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28977491

ABSTRACT

Cancer genome sequencing has implicated the cytosine deaminase activity of apolipoprotein B mRNA editing enzyme catalytic polypeptide-like (APOBEC) genes as an important source of mutations in diverse cancers, with APOBEC3B (A3B) expression especially correlated with such cancer mutations. To better understand the processes directing A3B over-expression in cancer, and possible therapeutic avenues for targeting A3B, we have investigated the regulation of A3B gene expression. Here, we show that A3B expression is inversely related to p53 status in different cancer types and demonstrate that this is due to a direct and pivotal role for p53 in repressing A3B expression. This occurs through the induction of p21 (CDKN1A) and the recruitment of the repressive DREAM complex to the A3B gene promoter, such that loss of p53 through mutation, or human papilloma virus-mediated inhibition, prevents recruitment of the complex, thereby causing elevated A3B expression and cytosine deaminase activity in cancer cells. As p53 is frequently mutated in cancer, our findings provide a mechanism by which p53 loss can promote cancer mutagenesis.


Subject(s)
Cytidine Deaminase/genetics , Gene Expression Regulation, Neoplastic , Minor Histocompatibility Antigens/genetics , Tumor Suppressor Protein p53/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytidine Deaminase/metabolism , HCT116 Cells , Humans , Immunoblotting , Minor Histocompatibility Antigens/metabolism , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism
5.
Cancer Lett ; 400: 194-202, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28259822

ABSTRACT

Although epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are effective for non-small cell lung cancer (NSCLC) patients with EGFR mutations, almost all these patients will eventually develop acquired resistance to EGFR-TKI. However, the molecular mechanisms responsible for gefitinib resistance remain still not fully understood. Here, we report that elevated DDX17 levels are observed in gefitinib-resistant NSCLC cells than gefitinib-sensitive cells. Upregulation of DDX17 enhances the gefitinib resistance, whereas DDX17-silenced cells partially restore gefitinib sensitivity. Mechanistically, we demonstrate that DDX17 disassociates the E-cadherin/ß-catenin complex, resulting in ß-catenin nuclear translocation and subsequently augmenting the transcription of ß-catenin target genes. Moreover, we identify two nuclear localization signal (NLS) and four nuclear export signal (NES) sequences mediated DDX17 nucleocytoplasmic shuttling via an exportin/importin-dependent pathways. Interruption of dynamic nucleocytoplasmic shuttling of DDX17 impairs DDX17-mediating the activation of ß-catenin and acquired resistance in NSCLC cells. In conclusion, our findings reveal a novel and important mechanism by which DDX17 contributes to acquired gefitinib resistance through exportin/importin-dependent cytoplasmic shuttling and followed by activation of ß-catenin, and DDX17 inhibition may be a promising strategy to overcome acquired resistance of gefitinib in NSCLC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , DEAD-box RNA Helicases/metabolism , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , beta Catenin/metabolism , A549 Cells , Active Transport, Cell Nucleus , Antigens, CD , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , DEAD-box RNA Helicases/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gefitinib , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Nuclear Export Signals , Nuclear Localization Signals , Protein Binding , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection
6.
Elife ; 52016 09 15.
Article in English | MEDLINE | ID: mdl-27630122

ABSTRACT

TP53 is conventionally thought to prevent cancer formation and progression to metastasis, while mutant TP53 has transforming activities. However, in the clinic, TP53 mutation status does not accurately predict cancer progression. Here we report, based on clinical analysis corroborated with experimental data, that the p53 isoform Δ133p53ß promotes cancer cell invasion, regardless of TP53 mutation status. Δ133p53ß increases risk of cancer recurrence and death in breast cancer patients. Furthermore Δ133p53ß is critical to define invasiveness in a panel of breast and colon cell lines, expressing WT or mutant TP53. Endogenous mutant Δ133p53ß depletion prevents invasiveness without affecting mutant full-length p53 protein expression. Mechanistically WT and mutant Δ133p53ß induces EMT. Our findings provide explanations to 2 long-lasting and important clinical conundrums: how WT TP53 can promote cancer cell invasion and reciprocally why mutant TP53 gene does not systematically induce cancer progression.


Subject(s)
Breast Neoplasms/genetics , Colonic Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Tumor Suppressor Protein p53/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Colonic Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation , Neoplasm Invasiveness/genetics , Neoplasm Recurrence, Local/pathology , Protein Isoforms/genetics , Tumor Suppressor Protein p53/biosynthesis
7.
Biochim Biophys Acta ; 1859(10): 1299-305, 2016 10.
Article in English | MEDLINE | ID: mdl-27478153

ABSTRACT

MicroRNAs (miRNAs) are short (21-23nt long) RNAs that post-transcriptionally regulate gene expression in plants and animals. They are key regulators in all biological processes. In mammalian cells miRNAs are loaded into one of the four members of the Argonaute (Ago) protein family to form the RNA-induced silencing complex (RISC). RISCs inhibit the translation of mRNAs that share sequence complementarity with their loaded miRNAs. miRNA processing and miRNA-mediated gene regulation are highly regulated processes and involve many RNA-binding proteins as auxiliary factors. Here we show that the two RNA-binding proteins, p72 and KHSRP, both with known roles in promoting miRNA biogenesis, regulate the protein level of human Ago2 in transformed human cells. We determined that p72 and KHSRP influence Ago2 stability by regulating miRNA levels in the cell and that loss of p72/KHSRP results in a decrease of unloaded Ago2.


Subject(s)
Argonaute Proteins/genetics , DEAD-box RNA Helicases/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Argonaute Proteins/metabolism , Cell Line, Tumor , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Plasmids/chemistry , Plasmids/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Signal Transduction , Trans-Activators/metabolism , Transfection
8.
Clin Cancer Res ; 22(23): 5929-5938, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27301701

ABSTRACT

PURPOSE: CDK-activating kinase (CAK) is required for the regulation of the cell cycle and is a trimeric complex consisting of cyclin-dependent kinase 7 (CDK7), Cyclin H, and the accessory protein, MAT1. CDK7 also plays a critical role in regulating transcription, primarily by phosphorylating RNA polymerase II, as well as transcription factors such as estrogen receptor-α (ER). Deregulation of cell cycle and transcriptional control are general features of tumor cells, highlighting the potential for the use of CDK7 inhibitors as novel cancer therapeutics. EXPERIMENTAL DESIGN: mRNA and protein expression of CDK7 and its essential cofactors cyclin H and MAT1 were evaluated in breast cancer samples to determine if their levels are altered in cancer. Immunohistochemical staining of >900 breast cancers was used to determine the association with clinicopathologic features and patient outcome. RESULTS: We show that expressions of CDK7, cyclin H, and MAT1 are all closely linked at the mRNA and protein level, and their expression is elevated in breast cancer compared with the normal breast tissue. Intriguingly, CDK7 expression was inversely proportional to tumor grade and size, and outcome analysis showed an association between CAK levels and better outcome. Moreover, CDK7 expression was positively associated with ER expression and in particular with phosphorylation of ER at serine 118, a site important for ER transcriptional activity. CONCLUSIONS: Expressions of components of the CAK complex, CDK7, MAT1, and Cyclin H are elevated in breast cancer and correlate with ER. Like ER, CDK7 expression is inversely proportional to poor prognostic factors and survival. Clin Cancer Res; 22(23); 5929-38. ©2016 AACR.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cyclin H/genetics , Cyclin-Dependent Kinases/genetics , Gene Expression/genetics , Receptors, Estrogen/genetics , Adult , Cell Cycle Proteins , Female , Humans , Middle Aged , Phosphorylation/genetics , Prognosis , Signal Transduction/genetics , Transcription Factors , Transcription, Genetic/genetics , Cyclin-Dependent Kinase-Activating Kinase
9.
Sci Rep ; 6: 22848, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26947125

ABSTRACT

miRNAs are small RNAs that are key regulators of gene expression in eukaryotic organisms. The processing of miRNAs is regulated by structural characteristics of the RNA and is also tightly controlled by auxiliary protein factors. Among them, RNA binding proteins play crucial roles to facilitate or inhibit miRNA maturation and can be controlled in a cell, tissue and species-specific manners or in response to environmental stimuli. In this study we dissect the molecular mechanism that promotes the overexpression of miR-132 in mice over its related, co-transcribed and co-regulated miRNA, miR-212. We have shown that the loop structure of miR-132 is a key determinant for its efficient processing in cells. We have also identified a range of RNA binding proteins that recognize the loop of miR-132 and influence both miR-132 and miR-212 processing. The DEAD box helicase p72/DDX17 was identified as a factor that facilitates the specific processing of miR-132.


Subject(s)
DEAD-box RNA Helicases/metabolism , MicroRNAs/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Cell Line , Gene Expression Regulation , HeLa Cells , Humans , Mice , Models, Molecular , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional , Up-Regulation
11.
Nucleic Acids Res ; 44(2): 582-94, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26400164

ABSTRACT

Liver receptor homologue 1 (LRH-1) is an orphan nuclear receptor that has been implicated in the progression of breast, pancreatic and colorectal cancer (CRC). To determine mechanisms underlying growth promotion by LRH-1 in CRC, we undertook global expression profiling following siRNA-mediated LRH-1 knockdown in HCT116 cells, which require LRH-1 for growth and in HT29 cells, in which LRH-1 does not regulate growth. Interestingly, expression of the cell cycle inhibitor p21 (CDKN1A) was regulated by LRH-1 in HCT116 cells. p21 regulation was not observed in HT29 cells, where p53 is mutated. p53 dependence for the regulation of p21 by LRH-1 was confirmed by p53 knockdown with siRNA, while LRH-1-regulation of p21 was not evident in HCT116 cells where p53 had been deleted. We demonstrate that LRH-1-mediated p21 regulation in HCT116 cells does not involve altered p53 protein or phosphorylation, and we show that LRH-1 inhibits p53 recruitment to the p21 promoter, likely through a mechanism involving chromatin remodelling. Our study suggests an important role for LRH-1 in the growth of CRC cells that retain wild-type p53.


Subject(s)
Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation, Neoplastic , Receptors, Cytoplasmic and Nuclear/genetics , Tumor Suppressor Protein p53/genetics , Binding Sites , Chromatin Assembly and Disassembly , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Deletion , HCT116 Cells , HT29 Cells , Humans , Mutation , Organ Specificity , Phosphorylation , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
12.
Nature ; 528(7583): 517-22, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26675721

ABSTRACT

T helper 17 (TH17) lymphocytes protect mucosal barriers from infections, but also contribute to multiple chronic inflammatory diseases. Their differentiation is controlled by RORγt, a ligand-regulated nuclear receptor. Here we identify the RNA helicase DEAD-box protein 5 (DDX5) as a RORγt partner that coordinates transcription of selective TH17 genes, and is required for TH17-mediated inflammatory pathologies. Surprisingly, the ability of DDX5 to interact with RORγt and coactivate its targets depends on intrinsic RNA helicase activity and binding of a conserved nuclear long noncoding RNA (lncRNA), Rmrp, which is mutated in patients with cartilage-hair hypoplasia. A targeted Rmrp gene mutation in mice, corresponding to a gene mutation in cartilage-hair hypoplasia patients, altered lncRNA chromatin occupancy, and reduced the DDX5-RORγt interaction and RORγt target gene transcription. Elucidation of the link between Rmrp and the DDX5-RORγt complex reveals a role for RNA helicases and lncRNAs in tissue-specific transcriptional regulation, and provides new opportunities for therapeutic intervention in TH17-dependent diseases.


Subject(s)
DEAD-box RNA Helicases/metabolism , RNA, Long Noncoding/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , DEAD-box RNA Helicases/genetics , Female , Gene Expression Regulation/genetics , Hair/abnormalities , Hirschsprung Disease/genetics , Humans , Immunologic Deficiency Syndromes/genetics , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Organ Specificity , Osteochondrodysplasias/congenital , Osteochondrodysplasias/genetics , Primary Immunodeficiency Diseases , Protein Binding , RNA, Long Noncoding/genetics , Transcription, Genetic/genetics
13.
Cell Rep ; 13(1): 108-121, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26411678

ABSTRACT

Estrogen receptor α (ERα) is the key transcriptional driver in a large proportion of breast cancers. We report that APOBEC3B (A3B) is required for regulation of gene expression by ER and acts by causing C-to-U deamination at ER binding regions. We show that these C-to-U changes lead to the generation of DNA strand breaks through activation of base excision repair (BER) and to repair by non-homologous end-joining (NHEJ) pathways. We provide evidence that transient cytidine deamination by A3B aids chromatin modification and remodelling at the regulatory regions of ER target genes that promotes their expression. A3B expression is associated with poor patient survival in ER+ breast cancer, reinforcing the physiological significance of A3B for ER action.


Subject(s)
Breast Neoplasms/genetics , Cytidine Deaminase/genetics , Cytidine/metabolism , DNA End-Joining Repair , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cytidine Deaminase/antagonists & inhibitors , Cytidine Deaminase/metabolism , DNA/genetics , DNA/metabolism , DNA Damage , Deamination , Estrogen Receptor alpha/metabolism , Female , Humans , Minor Histocompatibility Antigens , Prognosis , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Transcription, Genetic , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
Oncotarget ; 6(25): 21685-703, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26280373

ABSTRACT

The Nuclear Receptor (NR) superfamily of transcription factors comprises 48 members, several of which have been implicated in breast cancer. Most important is estrogen receptor-α (ERα), which is a key therapeutic target. ERα action is facilitated by co-operativity with other NR and there is evidence that ERα function may be recapitulated by other NRs in ERα-negative breast cancer. In order to examine the inter-relationships between nuclear receptors, and to obtain evidence for previously unsuspected roles for any NRs, we undertook quantitative RT-PCR and bioinformatics analysis to examine their expression in breast cancer. While most NRs were expressed, bioinformatic analyses differentiated tumours into distinct prognostic groups that were validated by analyzing public microarray data sets. Although ERα and progesterone receptor were dominant in distinguishing prognostic groups, other NR strengthened these groups. Clustering analysis identified several family members with potential importance in breast cancer. Specifically, RORγ is identified as being co-expressed with ERα, whilst several NRs are preferentially expressed in ERα-negative disease, with TLX expression being prognostic in this subtype. Functional studies demonstrated the importance of TLX in regulating growth and invasion in ERα-negative breast cancer cells.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Receptors, Cytoplasmic and Nuclear/metabolism , Triple Negative Breast Neoplasms/metabolism , Breast Neoplasms/metabolism , Cell Nucleus/metabolism , Cluster Analysis , Computational Biology , Estrogen Receptor alpha/metabolism , Female , Humans , Neoplasm Invasiveness , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Oligonucleotide Array Sequence Analysis , Orphan Nuclear Receptors , Prognosis
15.
Methods Mol Biol ; 1259: 17-33, 2015.
Article in English | MEDLINE | ID: mdl-25579577

ABSTRACT

RNA helicases of the DEAD-box family are found in all eukaryotes, most bacteria and many archaea. They play important roles in rearranging RNA-RNA and RNA-protein interactions. DEAD-box proteins are ATP-dependent RNA binding proteins and RNA-dependent ATPases. The first helicases of this large family of proteins were described in the 1980s. Since then our perception of these proteins has dramatically changed. From bona fide helicases, they became RNA binding proteins that separate duplex RNAs, in a local manner, by binding and bending the target RNA. In the present review we describe some of the experiments that were important milestones in the life of DEAD-box proteins since their birth 25 years ago.


Subject(s)
DEAD-box RNA Helicases/metabolism , RNA Helicases/metabolism , Adenosine Triphosphatases/metabolism , Protein Binding , RNA/chemistry , RNA/metabolism
16.
Cell Cycle ; 13(9): 1413-23, 2014.
Article in English | MEDLINE | ID: mdl-24626184

ABSTRACT

p68 (DDX5) acts both as an ATP-dependent RNA helicase and as a transcriptional co-activator of several cancer-associated transcription factors, including the p53 tumor suppressor. p68 is aberrantly expressed in a high proportion of cancers, but the oncogenic drive for, or the consequences of, these expression changes remain unclear. Here we show that elevated p68 expression in a cohort of human breast cancers is associated significantly with elevated levels of the oncogenic protein kinase, Polo-like kinase-1 (PLK1). Patients expressing detectable levels of both p68 and PLK1 have a poor prognosis, but only if they also have mutation in the TP53 gene (encoding p53), suggesting that p68 can regulate PLK1 levels in a manner that is suppressed by p53. In support of this hypothesis, we show that p68 stimulates expression from the PLK1 promoter, and that silencing of endogenous p68 expression downregulates endogenous PLK1 gene expression. In the absence of functional p53, p68 stimulates the expression of PLK1 both at basal levels and in response to the clinically relevant drug, etoposide. In keeping with a role as a transcriptional activator/co-activator, chromatin immuno-precipitation analysis shows that p68 is associated with the PLK1 promoter, irrespective of the p53 status. However, its recruitment is stimulated by etoposide in cells lacking p53, suggesting that p53 can oppose association of p68 with the PLK1 promoter. These data provide a model in which p68 and p53 interplay regulates PLK1 expression, and which describes the behavior of these molecules, and the outcome of their interaction, in human breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle Proteins/genetics , DEAD-box RNA Helicases/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Adenosine Triphosphatases/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cohort Studies , Etoposide/pharmacology , Female , Humans , Middle Aged , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Polo-Like Kinase 1
18.
Biochim Biophys Acta ; 1829(8): 750-5, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23542735

ABSTRACT

DEAD-box proteins represent the largest family of RNA helicases, present in all three kingdoms of life. They are involved in a variety of processes involving RNA metabolism and in some instances also in processes that use guide RNAs. Since their first descriptions in the late 1980s, the perception of their molecular activities has dramatically changed. At the time when only eight proteins with 9 conserved motifs constituted the DEAD-box protein family, it was the biochemical characterization of mammalian eIF4A that first suggested a local unwinding activity. This was confirmed in vitro using partially double stranded RNA substrates with the unexpected result of a bidirectional unwinding activity. A real change of paradigm from the classical helicase activity to localized RNA unwinding occurred with the publication of the vasa•RNA structure with a bend in the RNA substrate and the insightful work from several laboratories demonstrating local unwinding without translocation. Finally, elegant work on the exon-junction complex revealed how DEAD-box proteins can bind to RNA to serve as clamps to function as nucleation centers to form RNP complexes. This article is part of a Special Issue entitled: The Biology of RNA helicases - Modulation for life.


Subject(s)
DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , RNA/genetics , RNA/metabolism , Animals , Bacteria/enzymology , Bacteria/genetics , Bacteria/metabolism , DEAD-box RNA Helicases/chemistry , Humans , Models, Molecular , RNA/chemistry
19.
Biochim Biophys Acta ; 1829(8): 756-63, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23523990

ABSTRACT

Members of the DEAD box family of RNA helicases, which are characterised by the presence of twelve conserved motifs (including the signature D-E-A-D motif) within a structurally conserved 'helicase' core, are involved in all aspects of RNA metabolism. Apart from unwinding RNA duplexes, which established these proteins as RNA helicases, DEAD box proteins have been shown to also catalyse RNA annealing and to displace proteins from RNA. DEAD box proteins generally act as components of large multi-protein complexes and it is thought that interactions, via their divergent N- and C-terminal extensions, with other factors in the complexes may be responsible for the many different functions attributed to these proteins. In addition to their established crucial roles in the manipulation of RNA structure, it is becoming increasingly clear that several members of the DEAD box family act as regulators of transcription. In this review I shall focus on DDX5 (p68) and the highly related DDX17 (p72), two proteins for which there is a large body of evidence demonstrating that they function in transcriptional regulation. This article is part of a Special Issue entitled: The Biology of RNA helicases - Modulation for life.


Subject(s)
DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , RNA/genetics , RNA/metabolism , Animals , Humans , Transcription, Genetic
20.
RNA Biol ; 10(1): 121-32, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23353573

ABSTRACT

Members of the DEAD box family of RNA helicases are known to be involved in most cellular processes that require manipulation of RNA structure and, in many cases, exhibit other functions in addition to their established ATP-dependent RNA helicase activities. They thus play critical roles in cellular metabolism and in many cases have been implicated in cellular proliferation and/or neoplastic transformation. These proteins generally act as components of multi-protein complexes; therefore their precise role is likely to be influenced by their interacting partners and to be highly context-dependent. This may also provide an explanation for the sometimes conflicting reports suggesting that DEAD box proteins have both pro- and anti-proliferative roles in cancer.


Subject(s)
DEAD-box RNA Helicases/metabolism , Neoplasms/metabolism , Animals , DEAD-box RNA Helicases/classification , DEAD-box RNA Helicases/genetics , Humans , Neoplasms/genetics , RNA/genetics , RNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...