Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 289(48): 33629-43, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25301952

ABSTRACT

The cellular response to mitogens is tightly regulated via transcriptional and post-transcriptional mechanisms to rapidly induce genes that promote proliferation and efficiently attenuate their expression to prevent malignant growth. RNase L is an endoribonuclease that mediates diverse antiproliferative activities, and tristetraprolin (TTP) is a mitogen-induced RNA-binding protein that directs the decay of proliferation-stimulatory mRNAs. In light of their roles as endogenous proliferative constraints, we examined the mechanisms and functional interactions of RNase L and TTP to attenuate a mitogenic response. Mitogen stimulation of RNase L-deficient cells significantly increased TTP transcription and the induction of other mitogen-induced mRNAs. This regulation corresponded with elevated expression of serum-response factor (SRF), a master regulator of mitogen-induced transcription. RNase L destabilized the SRF transcript and formed a complex with SRF mRNA in cells providing a mechanism by which RNase L down-regulates SRF-induced genes. TTP and RNase L proteins interacted in cells suggesting that RNase L is directed to cleave TTP-bound RNAs as a mechanism of substrate specificity. Consistent with their concerted function in RNA turnover, the absence of either RNase L or TTP stabilized SRF mRNA, and a subset of established TTP targets was also regulated by RNase L. RNase L deficiency enhanced mitogen-induced proliferation demonstrating its functional role in limiting the mitogenic response. Our findings support a model of feedback regulation in which RNase L and TTP target SRF mRNA and SRF-induced transcripts. Accordingly, meta-analysis revealed an enrichment of RNase L and TTP targets among SRF-regulated genes suggesting that the RNase L/TTP axis represents a viable target to inhibit SRF-driven proliferation in neoplastic diseases.


Subject(s)
Cell Proliferation/drug effects , Endoribonucleases/metabolism , Gene Expression Regulation/drug effects , Mitogens/pharmacology , RNA Stability/drug effects , Transcription, Genetic/drug effects , Animals , Cell Proliferation/physiology , Gene Expression Regulation/physiology , HEK293 Cells , HeLa Cells , Humans , Mice , Models, Biological , RNA Stability/physiology , Transcription, Genetic/physiology , Tristetraprolin/metabolism
2.
J Interferon Cytokine Res ; 34(4): 275-88, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24697205

ABSTRACT

RNase-L is a mediator of type 1 interferon-induced antiviral activity that has diverse and critical cellular roles, including the regulation of cell proliferation, differentiation, senescence and apoptosis, tumorigenesis, and the control of the innate immune response. Although RNase-L was originally shown to mediate the endonucleolytic cleavage of both viral and ribosomal RNAs in response to infection, more recent evidence indicates that RNase-L also functions in the regulation of cellular mRNAs as an important mechanism by which it exerts its diverse biological functions. Despite this growing body of work, many questions remain regarding the roles of mRNAs as RNase-L substrates. This review will survey known and putative mRNA substrates of RNase-L, propose mechanisms by which it may selectively cleave these transcripts, and postulate future clinical applications.


Subject(s)
Endoribonucleases/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Virus Diseases/immunology , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Endoribonucleases/genetics , Female , Gene Expression Regulation , Humans , Immunity, Innate , Interferon Type I/immunology , Male , RNA, Viral/genetics , Substrate Specificity , Virus Diseases/genetics
3.
Inflamm Bowel Dis ; 19(6): 1295-305, 2013 May.
Article in English | MEDLINE | ID: mdl-23567782

ABSTRACT

BACKGROUND: The endoribonuclease RNase-L is a type-I interferon (IFN)-regulated component of the innate immune response that functions in antiviral, antibacterial, and antiproliferative activities. RNase-L produces RNA agonists of RIG-I-like receptors, sensors of cytosolic pathogen-associated RNAs that induce cytokines including IFN-ß. IFN-ß and RIG-I-like receptors signaling mediate protective responses against experimental colitis and colitis-associated cancer and contribute to gastrointestinal homeostasis. Therefore, we investigated a role for RNase-L in murine colitis and colitis-associated cancer and its association with RIG-I-like receptors signaling in response to bacterial RNA. METHODS: Colitis was induced in wild type-deficient and RNase-L-deficient mice (RNase-L⁻/⁻) by administration of dextran sulfate sodium (DSS). Colitis-associated cancer was induced by DSS and azoxymethane (AOM). Histological analysis and immunohistochemistry were performed on colon tissue to analyze immune cell infiltration and tissue damage after induction of colitis. Expression of cytokines was measured by quantitative real-time-PCR and ELISA. RESULTS: DSS-treated RNase-L⁻/⁻ mice exhibited a significantly higher clinical score, delayed leukocyte infiltration, reduced expression of IFN-ß, tumor necrosis factor α, interleukin-1ß, and interleukin-18 at early times post-DSS exposure, and increased mortality as compared with wild-type mice. DSS/AOM-treated RNase-L⁻/⁻ mice displayed an increased tumor burden. Bacterial RNA triggered IFN-ß production in an RNase-L-dependent manner and provided a potential mechanism by which RNase-L contributes to the gastrointestinal immune response to microbiota and protects against experimental colitis and colitis-associated cancer. CONCLUSIONS: RNase-L promotes the innate immune response to intestinal damage and ameliorates murine colitis and colitis-associated cancer. The RNase-L-dependent production of IFN-ß stimulated by bacterial RNA may be a mechanism to protect against gastrointestinal inflammatory disease.


Subject(s)
Colitis/complications , Colonic Neoplasms/etiology , Disease Models, Animal , Endoribonucleases/physiology , Immunity, Innate/immunology , Interferon Type I/metabolism , Animals , Azoxymethane/toxicity , Blotting, Western , Carcinogens/toxicity , Colitis/chemically induced , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytokines/genetics , Cytokines/metabolism , Dextran Sulfate/toxicity , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
4.
PLoS One ; 7(3): e33194, 2012.
Article in English | MEDLINE | ID: mdl-22413002

ABSTRACT

The serine/threonine kinase Pim-1 directs selected signaling events that promote cell growth and survival and is overexpressed in diverse human cancers. Pim-1 expression is tightly controlled through multiple mechanisms, including regulation of mRNA turnover. In several cultured cell models, mitogenic stimulation rapidly induced and stabilized PIM1 mRNA, however, vigorous destabilization 4-6 hours later helped restore basal expression levels. Acceleration of PIM1 mRNA turnover coincided with accumulation of tristetraprolin (TTP), an mRNA-destabilizing protein that targets transcripts containing AU-rich elements. TTP binds PIM1 mRNA in cells, and suppresses its expression by accelerating mRNA decay. Reporter mRNA decay assays localized the TTP-regulated mRNA decay element to a discrete AU-rich sequence in the distal 3'-untranslated region that binds TTP. These data suggest that coordinated stimulation of TTP and PIM1 expression limits the magnitude and duration of PIM1 mRNA accumulation by accelerating its degradation as TTP protein levels increase. Consistent with this model, PIM1 and TTP mRNA levels were well correlated across selected human tissue panels, and PIM1 mRNA was induced to significantly higher levels in mitogen-stimulated fibroblasts from TTP-deficient mice. Together, these data support a model whereby induction of TTP mediates a negative feedback circuit to limit expression of selected mitogen-activated genes.


Subject(s)
Gene Expression Regulation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-pim-1/metabolism , Tristetraprolin/genetics , Tristetraprolin/metabolism , 3' Untranslated Regions , AT Rich Sequence , Animals , Base Sequence , Cell Culture Techniques , Cell Line , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Knockout , Mitogens/pharmacology , Molecular Sequence Data , Organ Specificity/genetics , Proto-Oncogene Proteins c-pim-1/genetics , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements , Transcription, Genetic/drug effects
5.
Ageing Res Rev ; 11(4): 473-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22387927

ABSTRACT

Cancer and senescence are both complex transformative processes that dramatically alter many features of cell physiology and their interactions with surrounding tissues. Developing the wide range of cellular features characteristic of these conditions requires profound alterations in global gene expression patterns, which can be achieved by suppressing, activating, or uncoupling cellular gene regulatory pathways. Many genes associated with the initiation and development of tumors are regulated at the level of mRNA decay, frequently through the activity of AU-rich mRNA-destabilizing elements (AREs) located in their 3'-untranslated regions. As such, cellular factors that recognize and control the decay of ARE-containing mRNAs can influence tumorigenic or senescent phenotypes mediated by products of these transcripts. In this review, we discuss evidence showing how suppressed expression and/or activity of the ARE-binding protein tristetraprolin (TTP) can contribute to these processes. Next, we outline current findings linking TTP suppression to exacerbation of individual tumorigenic phenotypes, and the roles of specific TTP substrate mRNAs in mediating these effects. Finally, we survey potential mechanisms that cells may employ to suppress TTP expression in cancer, and propose potential diagnostic and therapeutic strategies that may exploit the relationship between TTP expression and tumor progression or senescence.


Subject(s)
Aging/genetics , Cellular Senescence/genetics , Neoplasms/genetics , RNA Processing, Post-Transcriptional/genetics , Tristetraprolin/genetics , Aging/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasms/metabolism , Neoplasms/physiopathology , RNA Stability/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...