Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37162991

ABSTRACT

5-fluorouracil (5-FU) is a successful and broadly used anti-cancer therapeutic. A major mechanism of action of 5-FU is thought to be through thymidylate synthase (TYMS) inhibition resulting in dTTP depletion and activation of the DNA damage response. This suggests that 5-FU should synergize with other DNA damaging agents. However, we found that combinations of 5-FU and oxaliplatin or irinotecan failed to display any evidence of synergy in clinical trials, and resulted in sub-additive killing in a panel of colorectal cancer (CRC) cell lines. In seeking to understand this antagonism, we unexpectedly found that an RNA damage response during ribosome biogenesis dominates the drug's efficacy in tumor types for which 5-FU shows clinical benefit. 5-FU has an inherent bias for RNA incorporation, and blocking this greatly reduced drug-induced lethality, indicating that accumulation of damaged RNA is more deleterious than the lack of new RNA synthesis. Using 5-FU metabolites that specifically incorporate into either RNA or DNA revealed that CRC cell lines and patient-derived colorectal cancer organoids are inherently more sensitive to RNA damage. This difference held true in cell lines from other tissues in which 5-FU has shown clinical utility, whereas cell lines from tumor tissues that lack clinical 5-FU responsiveness typically showed greater sensitivity to the drug's DNA damage effects. Analysis of changes in the phosphoproteome and ubiquitinome shows RNA damage triggers the selective ubiquitination of multiple ribosomal proteins leading to autophagy-dependent rRNA catabolism and proteasome-dependent degradation of ubiquitinated ribosome proteins. Further, RNA damage response to 5-FU is selectively enhanced by compounds that promote ribosome biogenesis, such as KDM2A inhibitors. These results demonstrate the presence of a strong RNA damage response linked to apoptotic cell death, with clear utility of combinatorially targeting this response in cancer therapy.

2.
Front Mol Biosci ; 10: 1148933, 2023.
Article in English | MEDLINE | ID: mdl-37091863

ABSTRACT

14-3-3 proteins play critical roles in controlling multiple aspects of the cellular response to stress and DNA damage including regulation of metabolism, cell cycle progression, cell migration, and apoptotic cell death by binding to protein substrates of basophilic protein kinases following their phosphorylation on specific serine/threonine residues. Although over 200 mammalian proteins that bind to 14-3-3 have been identified, largely through proteomic studies, in many cases the relevant protein kinase responsible for conferring 14-3-3-binding to these proteins is not known. To facilitate the identification of kinase-specific 14-3-3 clients, we developed a biochemical approach using high-density protein filter arrays and identified the translational regulatory molecule PABPC1 as a substrate for Chk1 and MAPKAP Kinase-2 (MK2) in vitro, and for MK2 in vivo, whose phosphorylation results in 14-3-3-binding. We identify Ser-470 on PABPC1 within the linker region connecting the RRM domains to the PABC domain as the critical 14-3-3-binding site, and demonstrate that loss of PABPC1 binding to 14-3-3 results in increased cell proliferation and decreased cell death in response to UV-induced DNA damage.

4.
Sci Signal ; 14(705): eabc4764, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34665642

ABSTRACT

Although immune checkpoint blockade (ICB) has strong clinical benefit for treating some tumor types, it fails in others, indicating a need for additional modalities to enhance the ICB effect. Here, we identified one such modality by using DNA damage to create a live, injured tumor cell adjuvant. Using an optimized ex vivo coculture system, we found that treating tumor cells with specific concentrations of etoposide, mitoxantrone, or doxorubicin markedly enhanced dendritic cell­mediated T cell activation. These immune-enhancing effects of DNA damage did not correlate with immunogenic cell death markers or with the extent of apoptosis or necroptosis; instead, these effects were mediated by live injured cells with activation of the DNA-PK, ATR, NF-κB, p38 MAPK, and RIPK1 signaling pathways. In mice, intratumoral injection of ex vivo etoposide­treated tumor cells in combination with systemic ICB (by anti-PD-1 and anti-CTLA4 antibodies) increased the number of intratumoral CD103+ dendritic cells and circulating tumor-antigen­specific CD8+ T cells, decreased tumor growth, and improved survival. These effects were absent in Batf3−/− mice and in mice in which the DNA-damaging drug was injected directly into the tumor, due to DNA damage in the immune cells. The combination treatment induced complete tumor regression in a subset of mice that were then able to reject tumor rechallenge, indicating that the injured cell adjuvant treatment induced durable antitumor immunological memory. These results provide a strategy for enhancing the efficacy of immune checkpoint inhibition in tumor types that do not respond to this treatment modality by itself.


Subject(s)
DNA Damage
5.
Nat Commun ; 11(1): 4124, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32807787

ABSTRACT

In response to DNA damage, a synthetic lethal relationship exists between the cell cycle checkpoint kinase MK2 and the tumor suppressor p53. Here, we describe the concept of augmented synthetic lethality (ASL): depletion of a third gene product enhances a pre-existing synthetic lethal combination. We show that loss of the DNA repair protein XPA markedly augments the synthetic lethality between MK2 and p53, enhancing anti-tumor responses alone and in combination with cisplatin chemotherapy. Delivery of siRNA-peptide nanoplexes co-targeting MK2 and XPA to pre-existing p53-deficient tumors in a highly aggressive, immunocompetent mouse model of lung adenocarcinoma improves long-term survival and cisplatin response beyond those of the synthetic lethal p53 mutant/MK2 combination alone. These findings establish a mechanism for co-targeting DNA damage-induced cell cycle checkpoints in combination with repair of cisplatin-DNA lesions in vivo using RNAi nanocarriers, and motivate further exploration of ASL as a generalized strategy to improve cancer treatment.


Subject(s)
Cell Cycle Checkpoints/physiology , DNA Repair/physiology , Animals , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , HCT116 Cells , Humans , Immunoblotting , Mice , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nanomedicine/methods , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
6.
Cell Rep ; 32(8): 108064, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32846122

ABSTRACT

RNA-binding proteins (RBPs) play critical roles in regulating gene expression by modulating splicing, RNA stability, and protein translation. Stimulus-induced alterations in RBP function contribute to global changes in gene expression, but identifying which RBPs are responsible for the observed changes remains an unmet need. Here, we present Transite, a computational approach that systematically infers RBPs influencing gene expression through changes in RNA stability and degradation. As a proof of principle, we apply Transite to RNA expression data from human patients with non-small-cell lung cancer whose tumors were sampled at diagnosis or after recurrence following treatment with platinum-based chemotherapy. Transite implicates known RBP regulators of the DNA damage response and identifies hnRNPC as a new modulator of chemotherapeutic resistance, which we subsequently validated experimentally. Transite serves as a framework for the identification of RBPs that drive cell-state transitions and adds additional value to the vast collection of publicly available gene expression datasets.


Subject(s)
DNA Damage/genetics , Gene Expression/genetics , RNA-Binding Proteins/metabolism , Humans
7.
Nat Commun ; 11(1): 4083, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796829

ABSTRACT

Proper chromatin function and maintenance of genomic stability depends on spatiotemporal coordination between the transcription and replication machinery. Loss of this coordination can lead to DNA damage from increased transcription-replication collision events. We report that deregulated transcription following BRD4 loss in cancer cells leads to the accumulation of RNA:DNA hybrids (R-loops) and collisions with the replication machinery causing replication stress and DNA damage. Whole genome BRD4 and γH2AX ChIP-Seq with R-loop IP qPCR reveals that BRD4 inhibition leads to accumulation of R-loops and DNA damage at a subset of known BDR4, JMJD6, and CHD4 co-regulated genes. Interference with BRD4 function causes transcriptional downregulation of the DNA damage response protein TopBP1, resulting in failure to activate the ATR-Chk1 pathway despite increased replication stress, leading to apoptotic cell death in S-phase and mitotic catastrophe. These findings demonstrate that inhibition of BRD4 induces transcription-replication conflicts, DNA damage, and cell death in oncogenic cells.


Subject(s)
Cell Cycle Proteins/pharmacology , DNA Damage/drug effects , DNA Replication/drug effects , R-Loop Structures/drug effects , Transcription Factors/pharmacology , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/metabolism , Chromatin , DNA-Binding Proteins , Genomic Instability , HeLa Cells , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Neoplasms/therapy , Nuclear Proteins/metabolism , S Phase , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
8.
Semin Thromb Hemost ; 46(2): 183-188, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32160643

ABSTRACT

Plasmin generation in trauma patients has wide-ranging effects, from breakdown of clots to remodeling the extracellular matrix. An evolving recognition of plasmin as a critical effector molecule in various inflammatory signals and pathways has rendered the study of plasmin(ogen) and its regulation by upstream activators and downstream targets and inhibitors key to understanding the inflammatory responses to trauma. Tranexamic acid, a widely available lysine analogue medication on the World Health Organization's list of essential medicines, has rapidly become one of the most commonly implemented adjunct treatments for bleeding after traumatic injury in clinical practice. In this article, we review the effects, both anti- and proinflammatory, of tranexamic acid, with a focus on the injured trauma patient.


Subject(s)
Antifibrinolytic Agents/therapeutic use , Fibrinolysis/drug effects , Tranexamic Acid/therapeutic use , Antifibrinolytic Agents/pharmacology , Humans , Signal Transduction , Tranexamic Acid/pharmacology
9.
Front Immunol ; 11: 607891, 2020.
Article in English | MEDLINE | ID: mdl-33708191

ABSTRACT

Chronic inflammation increases the risk for colorectal cancer through a variety of mechanisms involving the tumor microenvironment. MAPK-activated protein kinase 2 (MK2), a major effector of the p38 MAPK stress and DNA damage response signaling pathway, and a critical regulator of pro-inflammatory cytokine production, has been identified as a key contributor to colon tumorigenesis under conditions of chronic inflammation. We have previously described how genetic inactivation of MK2 in an inflammatory model of colon cancer results in delayed tumor progression, decreased tumor angiogenesis, and impaired macrophage differentiation into a pro-tumorigenic M2-like state. The molecular mechanism responsible for the impaired angiogenesis and tumor progression, however, has remained contentious and poorly defined. Here, using RNA expression analysis, assays of angiogenesis factors, genetic models, in vivo macrophage depletion and reconstitution of macrophage MK2 function using adoptive cell transfer, we demonstrate that MK2 activity in macrophages is necessary and sufficient for tumor angiogenesis during inflammation-induced cancer progression. We identify a critical and previously unappreciated role for MK2-dependent regulation of the well-known pro-angiogenesis factor CXCL-12/SDF-1 secreted by tumor associated-macrophages, in addition to MK2-dependent regulation of Serpin-E1/PAI-1 by several cell types within the tumor microenvironment.


Subject(s)
Angiogenic Proteins/metabolism , Colitis-Associated Neoplasms/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Neovascularization, Pathologic , Protein Serine-Threonine Kinases/metabolism , Tumor-Associated Macrophages/enzymology , Adoptive Transfer , Angiogenic Proteins/genetics , Animals , Cells, Cultured , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/pathology , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Transcription, Genetic , Tumor Microenvironment , Tumor-Associated Macrophages/transplantation
10.
Mol Cell Oncol ; 8(1): 1848233, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33553604

ABSTRACT

R-loops are intermediate structures of transcription that can accumulate when transcriptional elongation is blocked by inhibiting BRD4. In normal cells, R-loop persistence suppresses firing of adjacent replication origins. This control is lost in a subset of cancer cells, where BRD4 inhibition results in R-loop accumulation, leading to transcription-replication collisions and DNA double-strand breaks during S-phase, followed by cell death. This finding sheds new light on the mechanisms by which BRD4 inhibitors function as cancer therapies, and indicates that targeting other cellular events to cause R-loop accumulation may be useful for cancer treatment.

11.
J Trauma Acute Care Surg ; 86(1): 101-107, 2019 01.
Article in English | MEDLINE | ID: mdl-30575685

ABSTRACT

BACKGROUND: Both tissue plasminogen activator (tPA) in the circulation and urokinase (uPA) in tissues cleave plasminogen (PLG) to plasmin to promote clot lysis. Tranexamic acid (TXA) blocks both the tPA-dependent generation of plasmin on blood clots as well as active plasmin binding to polymerized fibrin, and is commonly administered for bleeding in trauma to limit fibrinolysis. In addition to lysing clots, however, active plasmin also cleaves complement proteins, potentially enhancing inflammation. Because TXA does not block uPA-dependent plasmin generation from PLG and instead augments it, we hypothesized that administration of TXA could enhance or inhibit proinflammatory C5a formation in a PLG activator-dependent manner. METHODS: Citrate platelet-poor plasma (PPP) and PPP depleted of complement protein C3 or PLG were obtained from healthy donors and commercial sources. Platelet-poor plasma was treated ex vivo with or without TXA and either with or without tPA or with or without uPA. Clotting was then induced by calcium and thrombin in clotted PPP experiments, while unclotted PPP experiments were treated with vehicle controls. C5a levels were measured via enzyme-linked immunosorbent assay. Data were expressed as mean ± SEM. RESULTS: Plasmin-mediated fibrinolysis by tPA in clotted PPP led to an approximately threefold increase in C5a production (p < 0.0001), which was significantly inhibited by TXA (p < 0.001). Paradoxically, when fibrinolysis was induced by uPA, TXA treatment led to further increases in C5a production beyond uPA alone (p < 0.0001). Furthermore, clotting was not required for C5a generation from uPA + TXA. C3 depletion had no effect on C5a production, while depletion of PLG eliminated it. CONCLUSIONS: Tranexamic acid administration can have proinflammatory or anti-inflammatory effects through regulating C5a generation by plasmin, depending on the predominating PLG activator. Tranexamic acid may cause significant inflammatory C5a elevations in injured tissues by augmenting uPA-mediated plasmin generation in a fibrin-independent manner. In contrast, TXA reduces C5a generation during tPA-mediated fibrinolysis that may reduce inflammatory responses. In vivo validation of these novel ex vivo findings is warranted and may have important clinical consequences.


Subject(s)
Anti-Inflammatory Agents/metabolism , Antifibrinolytic Agents/pharmacology , Complement C5a/metabolism , Inflammation Mediators/metabolism , Tranexamic Acid/pharmacology , Adult , Antifibrinolytic Agents/administration & dosage , Blood Coagulation/drug effects , Blood Coagulation/physiology , Complement C5a/drug effects , Female , Fibrinolysin/metabolism , Fibrinolysis/drug effects , Fibrinolysis/physiology , Hemorrhage/drug therapy , Hemorrhage/etiology , Humans , Male , Middle Aged , Plasminogen/drug effects , Plasminogen/metabolism , Signal Transduction/drug effects , Thrombin/metabolism , Tissue Plasminogen Activator/metabolism , Tranexamic Acid/administration & dosage , Urokinase-Type Plasminogen Activator/metabolism , Wounds and Injuries/complications
12.
Proc Natl Acad Sci U S A ; 115(18): E4236-E4244, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29666270

ABSTRACT

Chronic inflammation is a major risk factor for colorectal cancer. The p38/MAPKAP Kinase 2 (MK2) kinase axis controls the synthesis of proinflammatory cytokines that mediate both chronic inflammation and tumor progression. Blockade of this pathway has been previously reported to suppress inflammation and to prevent colorectal tumorigenesis in a mouse model of inflammation-driven colorectal cancer, by mechanisms that are still unclear. Here, using whole-animal and tissue-specific MK2 KO mice, we show that MK2 activity in the myeloid compartment promotes tumor progression by supporting tumor neoangiogenesis in vivo. Mechanistically, we demonstrate that MK2 promotes polarization of tumor-associated macrophages into protumorigenic, proangiogenic M2-like macrophages. We further confirmed our results in human cell lines, where MK2 chemical inhibition in macrophages impairs M2 polarization and M2 macrophage-induced angiogenesis. Together, this study provides a molecular and cellular mechanism for the protumorigenic function of MK2.


Subject(s)
Colorectal Neoplasms/blood supply , Colorectal Neoplasms/epidemiology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/enzymology , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Protein Serine-Threonine Kinases/genetics
13.
Bioeng Transl Med ; 3(1): 26-36, 2018 01.
Article in English | MEDLINE | ID: mdl-29376131

ABSTRACT

DNA damaging chemotherapy is a cornerstone of current front-line treatments for advanced ovarian cancer (OC). Despite the fact that a majority of these patients initially respond to therapy, most will relapse with chemo-resistant disease; therefore, adjuvant treatments that synergize with DNA-damaging chemotherapy could improve treatment outcomes and survival in patients with this deadly disease. Here, we report the development of a nanoscale peptide-nucleic acid complex that facilitates tumor-specific RNA interference therapy to chemosensitize advanced ovarian tumors to frontline platinum/taxane therapy. We found that the nanoplex-mediated silencing of the protein kinase, MK2, profoundly sensitized mouse models of high-grade serous OC to cytotoxic chemotherapy by blocking p38/MK2-dependent cell cycle checkpoint maintenance. Combined RNAi therapy improved overall survival by 37% compared with platinum/taxane chemotherapy alone and decreased metastatic spread to the lungs without observable toxic side effects. These findings suggest (a) that peptide nanoplexes can serve as safe and effective delivery vectors for siRNA and (b) that combined inhibition of MK2 could improve treatment outcomes in patients currently receiving frontline chemotherapy for advanced OC.

14.
Clin Cancer Res ; 21(19): 4410-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26034127

ABSTRACT

PURPOSE: Cross-talk and feedback between the RAS/RAF/MEK/ERK and PI3K/AKT/mTOR cell signaling pathways is critical for tumor initiation, maintenance, and adaptive resistance to targeted therapy in a variety of solid tumors. Combined blockade of these pathways-horizontal blockade-is a promising therapeutic strategy; however, compounded dose-limiting toxicity of free small molecule inhibitor combinations is a significant barrier to its clinical application. EXPERIMENTAL DESIGN: AZD6244 (selumetinib), an allosteric inhibitor of Mek1/2, and PX-866, a covalent inhibitor of PI3K, were co-encapsulated in a tumor-targeting nanoscale drug formulation-layer-by-layer (LbL) nanoparticles. Structure, size, and surface charge of the nanoscale formulations were characterized, in addition to in vitro cell entry, synergistic cell killing, and combined signal blockade. In vivo tumor targeting and therapy was investigated in breast tumor xenograft-bearing NCR nude mice by live animal fluorescence/bioluminescence imaging, Western blotting, serum cytokine analysis, and immunohistochemistry. RESULTS: Combined MAPK and PI3K axis blockade from the nanoscale formulations (160 ± 20 nm, -40 ± 1 mV) was synergistically toxic toward triple-negative breast (MDA-MB-231) and RAS-mutant lung tumor cells (KP7B) in vitro, effects that were further enhanced upon encapsulation. In vivo, systemically administered LbL nanoparticles preferentially targeted subcutaneous MDA-MB-231 tumor xenografts, simultaneously blocked tumor-specific phosphorylation of the terminal kinases Erk and Akt, and elicited significant disease stabilization in the absence of dose-limiting hepatotoxic effects observed from the free drug combination. Mice receiving untargeted, but dual drug-loaded nanoparticles exhibited progressive disease. CONCLUSIONS: Tumor-targeting nanoscale drug formulations could provide a more safe and effective means to synergistically block MAPK and PI3K in the clinic.


Subject(s)
Antineoplastic Agents/administration & dosage , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Nanoparticles , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/administration & dosage , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Nanoparticles/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
15.
Proc Natl Acad Sci U S A ; 107(28): 12499-504, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20616046

ABSTRACT

In recent years there have been major advances with respect to the identification of the protein components and mechanisms of microRNA (miRNA) mediated silencing. However, the complete and precise repertoire of components and mechanism(s) of action remain to be fully elucidated. Herein we reveal the identification of a family of three LIM domain-containing proteins, LIMD1, Ajuba and WTIP (Ajuba LIM proteins) as novel mammalian processing body (P-body) components, which highlight a novel mechanism of miRNA-mediated gene silencing. Furthermore, we reveal that LIMD1, Ajuba, and WTIP bind to Ago1/2, RCK, Dcp2, and eIF4E in vivo, that they are required for miRNA-mediated, but not siRNA-mediated gene silencing and that all three proteins bind to the mRNA 5' m(7)GTP cap-protein complex. Mechanistically, we propose the Ajuba LIM proteins interact with the m(7)GTP cap structure via a specific interaction with eIF4E that prevents 4EBP1 and eIF4G interaction. In addition, these LIM-domain proteins facilitate miRNA-mediated gene silencing by acting as an essential molecular link between the translationally inhibited eIF4E-m(7)GTP-5(')cap and Ago1/2 within the miRISC complex attached to the 3'-UTR of mRNA, creating an inhibitory closed-loop complex.


Subject(s)
Carrier Proteins/metabolism , Gene Silencing , MicroRNAs/metabolism , Animals , Carrier Proteins/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , Genes , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Mice , MicroRNAs/genetics , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
16.
Genes Dev ; 23(10): 1207-20, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19451221

ABSTRACT

UVB-induced lesions in mammalian cellular DNA can, through the process of mutagenesis, lead to carcinogenesis. However, eukaryotic cells have evolved complex mechanisms of genomic surveillance and DNA damage repair to counteract the effects of UVB radiation. We show that following UVB DNA damage, there is an overall inhibition of protein synthesis and translational reprogramming. This reprogramming allows selective synthesis of DDR proteins, such as ERCC1, ERCC5, DDB1, XPA, XPD, and OGG1 and relies on upstream ORFs in the 5' untranslated region of these mRNAs. Experiments with DNA-PKcs-deficient cell lines and a specific DNA-PKcs inhibitor demonstrate that both the general repression of mRNA translation and the preferential translation of specific mRNAs depend on DNA-PKcs activity, and therefore our data establish a link between a key DNA damage signaling component and protein synthesis.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA Repair Enzymes/metabolism , Polyribosomes/metabolism , Protein Biosynthesis/radiation effects , Protein Transport/radiation effects , RNA, Messenger/metabolism , Ultraviolet Rays , Cell Line, Tumor , DNA Damage/radiation effects , DNA Repair Enzymes/genetics , Gene Expression Regulation/radiation effects , HeLa Cells , Humans , Oligonucleotide Array Sequence Analysis , Open Reading Frames , Protein Biosynthesis/genetics
17.
Biochem Soc Trans ; 36(Pt 6): 1224-31, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19021530

ABSTRACT

miRNAs (microRNAs) are short non-coding RNAs that regulate gene expression post-transcriptionally. They generally bind to the 3'-UTR (untranslated region) of their target mRNAs and repress protein production by destabilizing the mRNA and translational silencing. The exact mechanism of miRNA-mediated translational repression is yet to be fully determined, but recent data from our laboratory have shown that the stage of translation which is inhibited by miRNAs is dependent upon the promoter used for transcribing the target mRNA. This review focuses on understanding how miRNA repression is operating in light of these findings and the questions that still remain.


Subject(s)
Gene Expression Regulation , MicroRNAs/metabolism , Animals , Humans , Peptides/metabolism , Promoter Regions, Genetic/genetics , Protein Biosynthesis , RNA Caps/metabolism
18.
Proc Natl Acad Sci U S A ; 105(26): 8866-71, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18579786

ABSTRACT

MicroRNAs (miRNAs) are noncoding RNAs that base pair imperfectly to homologous regions in target mRNAs and negatively influence the synthesis of the corresponding proteins. Repression is mediated by a number of mechanisms, one of which is the direct inhibition of protein synthesis. Surprisingly, previous studies have suggested that two mutually exclusive mechanisms exist, one acting at the initiation phase of protein synthesis and the other at a postinitiation event. Here, we resolve this apparent dichotomy by demonstrating that the promoter used to transcribe the mRNA influences the type of miRNA-mediated translational repression. Transcripts derived from the SV40 promoter that contain let-7 target sites in their 3' UTRs are repressed at the initiation stage of translation, whereas essentially identical mRNAs derived from the TK promoter are repressed at a postinitiation step. We also show that there is a miR-34 target site within the 3' UTR of c-myc mRNA and that promoter dependency is also true for this endogenous 3' UTR. Overall, these data establish a link between the nuclear history of an mRNA and the mechanism of miRNA-mediated translational regulation in the cytoplasm.


Subject(s)
MicroRNAs/genetics , Promoter Regions, Genetic/genetics , Protein Biosynthesis , 3' Untranslated Regions/genetics , Base Sequence , Cycloheximide/pharmacology , HeLa Cells , Humans , Molecular Sequence Data , Peptide Chain Initiation, Translational/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Mol Cell ; 23(3): 401-12, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16885029

ABSTRACT

During apoptosis there is a substantial reduction in the rate of protein synthesis, and yet some mRNAs avoid this translational inhibition. To determine the impact that receptor-mediated cell death has on the translational efficiency of a large number of mRNAs, translational profiling was performed on MCF7 cells treated with the apoptosis-inducing ligand TRAIL. Our data indicate that approximately 3% of mRNAs remain associated with the polysomes in apoptotic cells, and genes that are involved in transcription, chromatin modification/remodeling, and the Notch signaling pathway are particularly prevalent among the mRNAs that evade translational inhibition. Internal ribosome entry segments (IRESs) were identified in several of the mRNAs that remained associated with the polysomes during apoptosis, and, importantly, these IRESs functioned efficiently in apoptotic cells. Finally, the data showed that polypyrimidine tract binding protein (PTB, a known IRES trans-acting factor or ITAF) is pivotal in regulating the apoptotic process by controlling IRES function.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation , Polypyrimidine Tract-Binding Protein/physiology , Regulatory Sequences, Ribonucleic Acid/genetics , 5' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/genetics , Apoptosis Regulatory Proteins/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Membrane Glycoproteins/pharmacology , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribosomes/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...