Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Trends Genet ; 39(1): 74-88, 2023 01.
Article in English | MEDLINE | ID: mdl-36379743

ABSTRACT

Cancer was initially considered to be an exclusively genetic disease, but an interplay of dysregulated genetic and epigenetic mechanisms is now known to contribute to the cancer phenotype. More recently, chemical modifications of RNA molecules - the so-called epitranscriptome - have been found to regulate various aspects of RNA function and homeostasis. Specific enzymes, known as RNA-modifying proteins (RMPs), are responsible for depositing, removing, and reading chemical modifications in RNA. Intensive investigations in the epitranscriptomic field in recent years, in conjunction with great technological advances, have revealed the critical role of RNA modifications in regulating numerous cellular pathways. Furthermore, growing evidence has revealed that RNA modification machinery is often altered in human cancers, highlighting the enormous potential of RMPs as pharmacological targets or diagnostic markers.


Subject(s)
Neoplasms , RNA , Humans , RNA/genetics , RNA/metabolism , Epigenesis, Genetic/genetics , Neoplasms/genetics , Neoplasms/metabolism , Epigenomics
2.
Oncogene ; 39(17): 3443-3457, 2020 04.
Article in English | MEDLINE | ID: mdl-32108164

ABSTRACT

Perturbations in ribosome biogenesis have been associated with cancer. Such aberrations activate p53 through the RPL5/RPL11/5S rRNA complex-mediated inhibition of HDM2. Studies using animal models have suggested that this signaling pathway might constitute an important anticancer barrier. To gain a deeper insight into this issue in humans, here we analyze somatic mutations in RPL5 and RPL11 coding regions, reported in The Cancer Genome Atlas and International Cancer Genome Consortium databases. Using a combined computational and statistical approach, complemented by a range of biochemical and functional analyses in human cancer cell models, we demonstrate the existence of several mechanisms by which RPL5 mutations may impair wild-type p53 upregulation and ribosome biogenesis. Unexpectedly, the same approach provides only modest evidence for a similar role of RPL11, suggesting that RPL5 represents a preferred target during human tumorigenesis in cancers with wild-type p53. Furthermore, we find that several functional cancer-associated RPL5 somatic mutations occur as rare germline variants in general population. Our results shed light on the so-far enigmatic role of cancer-associated mutations in genes encoding ribosomal proteins, with implications for our understanding of the tumor suppressive role of the RPL5/RPL11/5S rRNA complex in human malignancies.


Subject(s)
Mutation , Neoplasms , Proto-Oncogene Proteins c-mdm2 , Ribosomal Proteins , Ribosomes , Tumor Suppressor Protein p53 , A549 Cells , Female , Humans , Male , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Ribosomal, 5S/genetics , RNA, Ribosomal, 5S/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Oncogene ; 37(18): 2351-2366, 2018 05.
Article in English | MEDLINE | ID: mdl-29429989

ABSTRACT

The nucleolus is the major site for synthesis of ribosomes, complex molecular machines that are responsible for protein synthesis. A wealth of research over the past 20 years has clearly indicated that both quantitative and qualitative alterations in ribosome biogenesis can drive the malignant phenotype via dysregulation of protein synthesis. However, numerous recent proteomic, genomic, and functional studies have implicated the nucleolus in the regulation of processes that are unrelated to ribosome biogenesis, including DNA-damage response, maintenance of genome stability and its spatial organization, epigenetic regulation, cell-cycle control, stress responses, senescence, global gene expression, as well as assembly or maturation of various ribonucleoprotein particles. In this review, the focus will be on features of rDNA genes, which make them highly vulnerable to DNA damage and intra- and interchromosomal recombination as well as built-in mechanisms that prevent and repair rDNA damage, and how dysregulation of this interplay affects genome-wide DNA stability, gene expression and the balance between euchromatin and heterochromatin. We will also present the most recent insights into how malfunction of these cellular processes may be a central driving force of human malignancies, and propose a promising new therapeutic approach for the treatment of cancer.


Subject(s)
Cell Nucleolus/physiology , Genomic Instability/physiology , Neoplasms/pathology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans
4.
Proc Natl Acad Sci U S A ; 114(4): E496-E505, 2017 01 24.
Article in English | MEDLINE | ID: mdl-27994142

ABSTRACT

The microRNA miR-504 targets TP53 mRNA encoding the p53 tumor suppressor. miR-504 resides within the fibroblast growth factor 13 (FGF13) gene, which is overexpressed in various cancers. We report that the FGF13 locus, comprising FGF13 and miR-504, is transcriptionally repressed by p53, defining an additional negative feedback loop in the p53 network. Furthermore, we show that FGF13 1A is a nucleolar protein that represses ribosomal RNA transcription and attenuates protein synthesis. Importantly, in cancer cells expressing high levels of FGF13, the depletion of FGF13 elicits increased proteostasis stress, associated with the accumulation of reactive oxygen species and apoptosis. Notably, stepwise neoplastic transformation is accompanied by a gradual increase in FGF13 expression and increased dependence on FGF13 for survival ("nononcogene addiction"). Moreover, FGF13 overexpression enables cells to cope more effectively with the stress elicited by oncogenic Ras protein. We propose that, in cells in which activated oncogenes drive excessive protein synthesis, FGF13 may favor survival by maintaining translation rates at a level compatible with the protein quality-control capacity of the cell. Thus, FGF13 may serve as an enabler, allowing cancer cells to evade proteostasis stress triggered by oncogene activation.


Subject(s)
Fibroblast Growth Factors/metabolism , Neoplasms/metabolism , Ribosomes/metabolism , Cell Line, Tumor , Cell Survival , Fibroblast Growth Factors/genetics , Humans , MicroRNAs/genetics , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics
5.
Semin Cancer Biol ; 37-38: 36-50, 2016 06.
Article in English | MEDLINE | ID: mdl-26721423

ABSTRACT

The nucleolus is the most prominent nuclear substructure assigned to produce ribosomes; molecular machines that are responsible for carrying out protein synthesis. To meet the increased demand for proteins during cell growth and proliferation the cell must increase protein synthetic capacity by upregulating ribosome biogenesis. While larger nucleolar size and number have been recognized as hallmark features of many tumor types, recent evidence has suggested that, in addition to overproduction of ribosomes, decreased ribosome biogenesis as well as qualitative changes in this process could also contribute to tumor initiation and cancer progression. Furthermore, the nucleolus has become the focus of intense attention for its involvement in processes that are clearly unrelated to ribosome biogenesis such as sensing and responding to endogenous and exogenous stressors, maintenance of genome stability, regulation of cell-cycle progression, cellular senescence, telomere function, chromatin structure, establishment of nuclear architecture, global regulation of gene expression and biogenesis of multiple ribonucleoprotein particles. The fact that dysregulation of many of these fundamental cellular processes may contribute to the malignant phenotype suggests that normal functioning of the nucleolus safeguards against the development of cancer and indicates its potential as a therapeutic approach. Here we review the recent advances made toward understanding these newly-recognized nucleolar functions and their roles in normal and cancer cells, and discuss possible future research directions.


Subject(s)
Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Neoplasms/pathology , Cell Nucleolus/pathology , Chromosome Segregation , DNA Damage , DNA, Ribosomal , Epigenesis, Genetic , Genomic Instability , Humans , Mitosis , RNA, Ribosomal/metabolism , Ribosomes/metabolism , Telomere
6.
Nat Cell Biol ; 15(8): 967-77, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23851489

ABSTRACT

The DNA damage response (DDR) pathway and ARF function as barriers to cancer development. Although commonly regarded as operating independently of each other, some studies proposed that ARF is positively regulated by the DDR. Contrary to either scenario, we found that in human oncogene-transformed and cancer cells, ATM suppressed ARF protein levels and activity in a transcription-independent manner. Mechanistically, ATM activated protein phosphatase 1, which antagonized Nek2-dependent phosphorylation of nucleophosmin (NPM), thereby liberating ARF from NPM and rendering it susceptible to degradation by the ULF E3-ubiquitin ligase. In human clinical samples, loss of ATM expression correlated with increased ARF levels and in xenograft and tissue culture models, inhibition of ATM stimulated the tumour-suppressive effects of ARF. These results provide insights into the functional interplay between the DDR and ARF anti-cancer barriers, with implications for tumorigenesis and treatment of advanced tumours.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Neoplasms/physiopathology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p14ARF/metabolism , ADP-Ribosylation Factor 1/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/metabolism , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Male , Mice , NIMA-Related Kinases , Neoplasms/enzymology , Neoplasms/pathology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Stability , Ribosomes/metabolism , Signal Transduction , Transplantation, Heterologous , Tumor Suppressor Protein p14ARF/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Proc Natl Acad Sci U S A ; 109(50): 20467-72, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23169665

ABSTRACT

Impairment of ribosomal biogenesis can activate the p53 protein independently of DNA damage. The ability of ribosomal proteins L5, L11, L23, L26, or S7 to bind Mdm2 and inhibit its ubiquitin ligase activity has been suggested as a critical step in p53 activation under these conditions. Here, we report that L5 and L11 are particularly important for this response. Whereas several other newly synthesized ribosomal proteins are degraded by proteasomes upon inhibition of Pol I activity by actinomycin D, L5 and L11 accumulate in the ribosome-free fraction where they bind to Mdm2. This selective accumulation of free L5 and L11 is due to their mutual protection from proteasomal degradation. Furthermore, the endogenous, newly synthesized L5 and L11 continue to be imported into nucleoli even after nucleolar disruption and colocalize with Mdm2, p53, and promyelocytic leukemia protein. This suggests that the disrupted nucleoli may provide a platform for L5- and L11-dependent p53 activation, implying a role for the nucleolus in p53 activation by ribosomal biogenesis stress. These findings may have important implications with respect to understanding the pathogenesis of diseases caused by impaired ribosome biogenesis.


Subject(s)
Ribosomal Proteins/metabolism , Ribosomes/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Animals , Base Sequence , Cell Line, Tumor , Cell Nucleolus/metabolism , Dactinomycin/pharmacology , Humans , Mice , Models, Biological , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/genetics , Stress, Physiological , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...