51
|
Changes in translational control after pro-apoptotic stress. Int J Mol Sci 2012; 14:177-90. [PMID: 23344027 PMCID: PMC3565257 DOI: 10.3390/ijms14010177] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 11/06/2012] [Accepted: 12/10/2012] [Indexed: 01/17/2023] Open
Abstract
In stressed cells, a general decrease in the rate of protein synthesis occurs due to modifications in the activity of translation initiation factors. Compelling data now indicate that these changes also permit a selective post-transcriptional expression of proteins necessary for either cell survival or completion of apoptosis when cells are exposed to severe or prolonged stress. In this review, we summarize the modifications that inhibit the activity of the main canonical translation initiation factors, and the data explaining how certain mRNAs encoding proteins involved in either cell survival or apoptosis can be selectively translated.
Collapse
|
52
|
Nucleotide composition of cellular internal ribosome entry sites defines dependence on NF45 and predicts a posttranscriptional mitotic regulon. Mol Cell Biol 2012; 33:307-18. [PMID: 23129811 DOI: 10.1128/mcb.00546-12] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The vast majority of cellular mRNAs initiate their translations through a well-defined mechanism of ribosome recruitment that occurs at the 5'-terminal 7-methylguanosine cap with the help of several canonical protein factors. A subset of cellular and viral mRNAs contain regulatory motifs in their 5' untranslated regions (UTRs), termed internal ribosome entry sites (IRES), that sidestep this canonical mode of initiation. On cellular mRNAs, this mechanism requires IRES trans-acting protein factors (ITAFs) that facilitate ribosome recruitment downstream of the cap. While several ITAFs and their target mRNAs have been empirically identified, the in silico prediction of targets has proved difficult. Here, we report that a high AU content (>60%) of the IRES-containing 5' UTRs serves as an excellent predictor of dependence on NF45, a recently identified ITAF. Moreover, we provide evidence that cells deficient in NF45 ITAF activity exhibit reduced IRES-mediated translation of X-linked inhibitor of apoptosis protein (XIAP) and cellular inhibitor of apoptosis protein 1 (cIAP1) mRNAs that, in turn, leads to dysregulated expression of their respective targets, survivin and cyclin E. This specific defect in IRES translation explains in part the cytokinesis impairment and senescence-like phenotype observed in HeLa cells expressing NF45 RNA interference (RNAi). This study uncovers a novel role for NF45 in regulating ploidy and highlights the importance of IRES-mediated translation in cellular homeostasis.
Collapse
|
53
|
Petz M, Them NCC, Huber H, Mikulits W. PDGF enhances IRES-mediated translation of Laminin B1 by cytoplasmic accumulation of La during epithelial to mesenchymal transition. Nucleic Acids Res 2012; 40:9738-49. [PMID: 22904067 PMCID: PMC3479205 DOI: 10.1093/nar/gks760] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The extracellular matrix protein Laminin B1 (LamB1) regulates tumor cell migration and invasion. Carcinoma cells acquire invasive properties by epithelial to mesenchymal transition (EMT), which is a fundamental step in dissemination of metastatic cells from the primary tumor. Recently, we showed that enhanced translation of LamB1 upon EMT of malignant hepatocytes is mediated by an internal ribosome entry site (IRES). We demonstrated that the IRES transacting factor La binds the minimal IRES motif and positively modulates IRES activity of LamB1. Here, we show that platelet-derived growth factor (PDGF) enhances IRES activity of LamB1 by the increasing cytoplasmic localization of La during EMT. Accordingly, cells expressing dominant negative PDGF receptor display reduced cytoplasmic accumulation of La and show no elevation of IRES activity or endogenous LamB1 levels after stimulation with PDGF. Furthermore, La-mediated regulation of LamB1 IRES activity predominantly depends on MAPK/ERK signaling downstream of PDGF. Notably, LamB1 expression is not significantly downregulated by the impairment of the translation initiation factor eIF4E. In vivo, knockdown of La associated with decreased LamB1 expression and reduced tumor growth. Together, these data suggest that PDGF is required for the cytoplasmic accumulation of La that triggers IRES-dependent translation of LamB1 during EMT.
Collapse
Affiliation(s)
- Michaela Petz
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | | | | | | |
Collapse
|
54
|
Zhang J, Dinh TN, Kappeler K, Tsaprailis G, Chen QM. La autoantigen mediates oxidant induced de novo Nrf2 protein translation. Mol Cell Proteomics 2012; 11:M111.015032. [PMID: 22207702 PMCID: PMC3433904 DOI: 10.1074/mcp.m111.015032] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 12/25/2011] [Indexed: 01/20/2023] Open
Abstract
Nrf2 gene encodes a transcription factor that regulates the expression of a cluster of antioxidant and detoxification genes. Recent works from our laboratory indicate that oxidative stress causes rapid de novo synthesis of Nrf2 protein. We have found that 5' Untranslated Region (5'UTR) of Nrf2 allows the mRNA to undergo an Internal Ribosomal Entry Site (IRES) mediated protein translation. Using liquid chromatography tandem MS, we have discovered that La/SSB protein bound to Nrf2 5'UTR in response to oxidative stress. In vitro RNA binding and in vivo ribonucleoprotein immunoprecipitation showed H(2)O(2) dose and time dependent increases of La/SSB binding to Nrf2 5'UTR. La/SSB protein translocated from the nuclei to cytoplasm and distributed in the perinuclear space in cells treated with H(2)O(2). Isolation of ribosomal fractions indicated that oxidants caused an association of La/SSB with ribosomes. Physical interaction of La/SSB with representative proteins from the small or large subunits of ribosomes was found to increase in cells responding to H(2)O(2) treatment. Knocking down La/SSB gene with siRNA prevented Nrf2 protein elevation or Nrf2 5'UTR activation by oxidants. In contrast, overexpression of La/SSB gene was able to enhance Nrf2 5'UTR activation and Nrf2 protein increase. Our data suggest that oxidants cause nuclear export of La/SSB protein and subsequent association of La/SSB with Nrf2 5'UTR and ribosomes. These events contribute to de novo Nrf2 protein translation because of oxidative stress.
Collapse
Affiliation(s)
- Jack Zhang
- From the ‡Department of Pharmacology, University of Arizona, College of Medicine, 1501 N. Campbell Ave, Tucson, Arizona 85724
| | - Thai Nho Dinh
- From the ‡Department of Pharmacology, University of Arizona, College of Medicine, 1501 N. Campbell Ave, Tucson, Arizona 85724
| | - Kyle Kappeler
- From the ‡Department of Pharmacology, University of Arizona, College of Medicine, 1501 N. Campbell Ave, Tucson, Arizona 85724
| | - George Tsaprailis
- §Center for Toxicology, College of Pharmacy, 1703 E. Mabel St Tucson, Arizona 85721
| | - Qin M. Chen
- From the ‡Department of Pharmacology, University of Arizona, College of Medicine, 1501 N. Campbell Ave, Tucson, Arizona 85724
| |
Collapse
|
55
|
Chen H, Hao Y, Wang L, Jia D, Ruan Y, Gu J. Sodium arsenite down-regulates the expression of X-linked inhibitor of apoptosis protein via translational and post-translational mechanisms in hepatocellular carcinoma. Biochem Biophys Res Commun 2012; 422:721-6. [PMID: 22627131 DOI: 10.1016/j.bbrc.2012.05.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 05/14/2012] [Indexed: 12/17/2022]
Abstract
X-linked inhibitor of apoptosis protein (XIAP) is a member of the inhibitors of apoptosis protein (IAP) family, and has been reported to exhibit elevated expression levels in hepatocellular carcinoma (HCC) and promote cell survival, metastasis and tumor recurrence. Targeting XIAP has proven effective for the inhibition of cancer cell proliferation and restoration of cancer cell chemosensitivity. Arsenic (or sodium arsenite) is a potent anti-tumor agent used to treat patients with acute promyelocytic leukemia (APL). Additionally, arsenic induces cell growth inhibition, cell cycle arrest and apoptosis in human HCC cells. In this study, we identified XIAP as a target for sodium arsenite-induced cytotoxicity in HCC. The exposure of HCC cell lines to sodium arsenite resulted in inhibition of XIAP expression in both a dose- and time-dependent manner. Sodium arsenite blocked the de novo XIAP synthesis and the activity of its internal ribosome entry site (IRES) element. Moreover, treatment with sodium arsenite decreased the protein stability of XIAP and induced its ubiquitin-proteasomal degradation. Overexpression of XIAP attenuated the pro-apoptotic effect of sodium arsenite in HCC. Taken together, our data demonstrate that sodium arsenite suppresses XIAP expression via translational and post-translational mechanisms in HCC.
Collapse
Affiliation(s)
- Hong Chen
- Gene Research Center, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
| | | | | | | | | | | |
Collapse
|
56
|
Versatility of RNA-Binding Proteins in Cancer. Comp Funct Genomics 2012; 2012:178525. [PMID: 22666083 PMCID: PMC3359819 DOI: 10.1155/2012/178525] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 02/28/2012] [Indexed: 01/22/2023] Open
Abstract
Posttranscriptional gene regulation is a rapid and efficient process to adjust the proteome of a cell to a changing environment. RNA-binding proteins (RBPs) are the master regulators of mRNA processing and translation and are often aberrantly expressed in cancer. In addition to well-studied transcription factors, RBPs are emerging as fundamental players in tumor development. RBPs and their mRNA targets form a complex network that plays a crucial role in tumorigenesis. This paper describes mechanisms by which RBPs influence the expression of well-known oncogenes, focusing on precise examples that illustrate the versatility of RBPs in posttranscriptional control of cancer development. RBPs appeared very early in evolution, and new RNA-binding domains and combinations of them were generated in more complex organisms. The identification of RBPs, their mRNA targets, and their mechanism of action have provided novel potential targets for cancer therapy.
Collapse
|
57
|
Licursi M, Komatsu Y, Pongnopparat T, Hirasawa K. Promotion of viral internal ribosomal entry site-mediated translation under amino acid starvation. J Gen Virol 2012; 93:951-962. [DOI: 10.1099/vir.0.040386-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cap-dependent and internal ribosomal entry site (IRES)-mediated translation are regulated differently within cells. Viral IRES-mediated translation often remains active when cellular cap-dependent translation is severely impaired under cellular stresses induced by virus infection. To investigate how cellular stresses influence the efficiency of viral IRES-mediated translation, we used a bicistronic luciferase reporter construct harbouring IRES elements from the following viruses: encephalomyocarditis virus (EMCV), foot-and-mouth disease virus (FMDV), hepatitis C virus (HCV) or human rhinovirus (HRV). NIH3T3 cells transfected with these bicistronic reporter constructs were subjected to different cellular stresses. Increased translation initiation was only observed under amino acid starvation when EMCV or FMDV IRES elements were present. To identify cellular mechanisms that promoted viral IRES-mediated translation, we tested the involvement of eukaryotic initiation factor 4E-binding protein (4E-BP), general control non-depressed 2 (GCN2) and eukaryotic initiation factor 2B (eIF2B), as these are known to be modulated under amino acid starvation. Knockdown of 4E-BP1 impaired the promotion of EMCV and FMDV IRES-mediated translation under amino acid starvation, whereas GCN2 and eIF2B were not involved. To further investigate how 4E-BP1 regulates translation initiated by EMCV and FMDV IRES elements, we used a phosphoinositide kinase-3 inhibitor (LY294002), an mTOR inhibitor (Torin1) or leucine starvation to mimic 4E-BP1 dephosphorylation induced by amino acid starvation. 4E-BP1 dephosphorylation induced by the treatments was not sufficient to promote viral IRES-mediated translation. These results suggest that 4E-BP1 regulates EMCV and FMDV IRES-mediated translation under amino acid starvation, but not via its dephosphorylation.
Collapse
Affiliation(s)
- Maria Licursi
- Division of Biomedical Science, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St John’s, NL A1B 3V6, Canada
| | - Yumiko Komatsu
- Division of Biomedical Science, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St John’s, NL A1B 3V6, Canada
| | - Theerawat Pongnopparat
- Division of Biomedical Science, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St John’s, NL A1B 3V6, Canada
| | - Kensuke Hirasawa
- Division of Biomedical Science, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St John’s, NL A1B 3V6, Canada
| |
Collapse
|
58
|
Tumor suppressor PDCD4 represses internal ribosome entry site-mediated translation of antiapoptotic proteins and is regulated by S6 kinase 2. Mol Cell Biol 2012; 32:1818-29. [PMID: 22431522 DOI: 10.1128/mcb.06317-11] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Apoptosis can be regulated by extracellular signals that are communicated by peptides such as fibroblast growth factor 2 (FGF-2) that have important roles in tumor cell proliferation. The prosurvival effects of FGF-2 are transduced by the activation of the ribosomal protein S6 kinase 2 (S6K2), which increases the expression of the antiapoptotic proteins X chromosome-linked Inhibitor of Apoptosis (XIAP) and Bcl-x(L). We now show that the FGF-2-S6K2 prosurvival signaling is mediated by the tumor suppressor programmed cell death 4 (PDCD4). We demonstrate that PDCD4 specifically binds to the internal ribosome entry site (IRES) elements of both the XIAP and Bcl-x(L) messenger RNAs and represses their translation by inhibiting the formation of the 48S translation initiation complex. Phosphorylation of PDCD4 by activated S6K2 leads to the degradation of PDCD4 and thus the subsequent derepression of XIAP and Bcl-x(L) translation. Our results identify PDCD4 as a specific repressor of the IRES-dependent translation of cellular mRNAs (such as XIAP and Bcl-x(L)) that mediate FGF-2-S6K2 prosurvival signaling and provide further insight into the role of PDCD4 in tumor suppression.
Collapse
|
59
|
Mojic M, Mijatovic S, Maksimovic-Ivanic D, Dinic S, Grdovic N, Miljkovic D, Stosic-Grujicic S, Tumino S, Fagone P, Mangano K, Zocca MB, Al-Abed Y, McCubrey JA, Nicoletti F. Saquinavir-NO-targeted S6 protein mediates sensitivity of androgen-dependent prostate cancer cells to TRAIL. Cell Cycle 2012; 11:1174-82. [PMID: 22370480 DOI: 10.4161/cc.11.6.19611] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We previously reported that the NO-modified form of HIV protease inhibitor Saquinavir (Saq) is a potent antitumoral agent efficient against numerous tumor cell lines in vitro and in vivo. In acute toxicity studies, doses of Saq-NO equivalent to DL100 of the parental drug were completely nontoxic. Beside direct effect on malignant cell growth, Saq-NO sensitizes certain type of cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated cell death. In this study, we evaluated the effects of Saq-NO on androgen-dependent prostate cancer LNCaP. Saq-NO inhibited both the growth of LNCaP cells in vitro and in xenograft models. Suppression of tumor growth was accompanied with cell cycle arrest in G 0/G 1 phase and established a persistent inhibition of proliferation. Furthermore, Saq-NO reverted sensitivity of LNCaP cells to TRAIL but not to TNF. Treatment of cells with Saq-NO induced transient upregulation of Akt and ERK1/2. This, however, did not represent the primary mode of action of Saq-NO, as elimination with specific inhibitors did not compromise the chemotherapic efficacy of the drug. However, permanent abrogation of phosphorylation of the S6 protein, which is the downstream target of both signaling pathways, was observed. Diminished S6 phosphorylation was associated with re-established sensitivity to TRAIL and reduction of X-linked inhibitor of apoptosis protein (XIAP). In summary, NO modification of Saq led to a new chemical entity with stronger and more pleiotropic antitumor activity than the parental drug.
Collapse
Affiliation(s)
- Marija Mojic
- Department of Immunology; Institute for Biological Research Sinisa Stankovic, Belgrade University, Belgrade, Serbia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Nakatake M, Monte-Mor B, Debili N, Casadevall N, Ribrag V, Solary E, Vainchenker W, Plo I. JAK2(V617F) negatively regulates p53 stabilization by enhancing MDM2 via La expression in myeloproliferative neoplasms. Oncogene 2012; 31:1323-33. [PMID: 21785463 DOI: 10.1038/onc.2011.313] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 06/17/2011] [Accepted: 06/18/2011] [Indexed: 01/10/2023]
Abstract
JAK2(V617F) is a gain of function mutation that promotes cytokine-independent growth of myeloid cells and accounts for a majority of myeloproliferative neoplasms (MPN). Mutations in p53 are rarely found in these diseases before acute leukemia transformation, but this does not rule out a role for p53 deregulation in disease progression. Using Ba/F3-EPOR cells and ex vivo cultured CD34(+) cells from MPN patients, we demonstrate that expression of JAK2(V617F) affected the p53 response to DNA damage. We show that E3 ubiquitin ligase MDM2 accumulated in these cells, due to an increased translation of MDM2 mRNA. Accumulation of the La autoantigen, which interacts with MDM2 mRNA and promotes its translation, was responsible for the increase in MDM2 protein level and the subsequent degradation of p53 after DNA damage. Downregulation of La protein or cell treatment with nutlin-3, a MDM2 antagonist, restored the p53 response to DNA damage and the cytokine-dependence of Ba/F3-EPOR-JAK2(V617F) cells. Altogether, these data indicate that the JAK2(V617F) mutation affects p53 response to DNA damage through the upregulation of La antigen and accumulation of MDM2. They also suggest that p53 functional inactivation accounts for the cytokine hypersensitivity of JAK2(V617F) MPN and might have a role in disease progression.
Collapse
|
61
|
Martino L, Pennell S, Kelly G, Bui TTT, Kotik-Kogan O, Smerdon SJ, Drake AF, Curry S, Conte MR. Analysis of the interaction with the hepatitis C virus mRNA reveals an alternative mode of RNA recognition by the human La protein. Nucleic Acids Res 2012; 40:1381-94. [PMID: 22009680 PMCID: PMC3273827 DOI: 10.1093/nar/gkr890] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 09/29/2011] [Accepted: 10/01/2011] [Indexed: 12/31/2022] Open
Abstract
Human La protein is an essential factor in the biology of both coding and non-coding RNAs. In the nucleus, La binds primarily to 3' oligoU containing RNAs, while in the cytoplasm La interacts with an array of different mRNAs lacking a 3' UUU(OH) trailer. An example of the latter is the binding of La to the IRES domain IV of the hepatitis C virus (HCV) RNA, which is associated with viral translation stimulation. By systematic biophysical investigations, we have found that La binds to domain IV using an RNA recognition that is quite distinct from its mode of binding to RNAs with a 3' UUU(OH) trailer: although the La motif and first RNA recognition motif (RRM1) are sufficient for high-affinity binding to 3' oligoU, recognition of HCV domain IV requires the La motif and RRM1 to work in concert with the atypical RRM2 which has not previously been shown to have a significant role in RNA binding. This new mode of binding does not appear sequence specific, but recognizes structural features of the RNA, in particular a double-stranded stem flanked by single-stranded extensions. These findings pave the way for a better understanding of the role of La in viral translation initiation.
Collapse
Affiliation(s)
- Luigi Martino
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, Division of Molecular Structure, MRC Biomedical NMR Centre, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, Pharmaceutical Science Division, King's College London, The Wolfson Wing, Guy's Campus, London SE1 1UL and Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Simon Pennell
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, Division of Molecular Structure, MRC Biomedical NMR Centre, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, Pharmaceutical Science Division, King's College London, The Wolfson Wing, Guy's Campus, London SE1 1UL and Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Geoff Kelly
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, Division of Molecular Structure, MRC Biomedical NMR Centre, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, Pharmaceutical Science Division, King's College London, The Wolfson Wing, Guy's Campus, London SE1 1UL and Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Tam T. T. Bui
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, Division of Molecular Structure, MRC Biomedical NMR Centre, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, Pharmaceutical Science Division, King's College London, The Wolfson Wing, Guy's Campus, London SE1 1UL and Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Olga Kotik-Kogan
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, Division of Molecular Structure, MRC Biomedical NMR Centre, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, Pharmaceutical Science Division, King's College London, The Wolfson Wing, Guy's Campus, London SE1 1UL and Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Stephen J. Smerdon
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, Division of Molecular Structure, MRC Biomedical NMR Centre, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, Pharmaceutical Science Division, King's College London, The Wolfson Wing, Guy's Campus, London SE1 1UL and Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Alex F. Drake
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, Division of Molecular Structure, MRC Biomedical NMR Centre, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, Pharmaceutical Science Division, King's College London, The Wolfson Wing, Guy's Campus, London SE1 1UL and Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Stephen Curry
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, Division of Molecular Structure, MRC Biomedical NMR Centre, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, Pharmaceutical Science Division, King's College London, The Wolfson Wing, Guy's Campus, London SE1 1UL and Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Maria R. Conte
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, Division of Molecular Structure, MRC Biomedical NMR Centre, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, Pharmaceutical Science Division, King's College London, The Wolfson Wing, Guy's Campus, London SE1 1UL and Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| |
Collapse
|
62
|
Petz M, Them N, Huber H, Beug H, Mikulits W. La enhances IRES-mediated translation of laminin B1 during malignant epithelial to mesenchymal transition. Nucleic Acids Res 2012; 40:290-302. [PMID: 21896617 PMCID: PMC3245933 DOI: 10.1093/nar/gkr717] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 07/26/2011] [Accepted: 08/21/2011] [Indexed: 12/13/2022] Open
Abstract
The majority of transcripts that harbor an internal ribosome entry site (IRES) are involved in cancer development via corresponding proteins. A crucial event in tumor progression referred to as epithelial to mesenchymal transition (EMT) allows carcinoma cells to acquire invasive properties. The translational activation of the extracellular matrix component laminin B1 (LamB1) during EMT has been recently reported suggesting an IRES-mediated mechanism. In this study, the IRES activity of LamB1 was determined by independent bicistronic reporter assays. Strong evidences exclude an impact of cryptic promoter or splice sites on IRES-driven translation of LamB1. Furthermore, no other LamB1 mRNA species arising from alternative transcription start sites or polyadenylation signals were detected that account for its translational control. Mapping of the LamB1 5'-untranslated region (UTR) revealed the minimal LamB1 IRES motif between -293 and -1 upstream of the start codon. Notably, RNA affinity purification showed that the La protein interacts with the LamB1 IRES. This interaction and its regulation during EMT were confirmed by ribonucleoprotein immunoprecipitation. In addition, La was able to positively modulate LamB1 IRES translation. In summary, these data indicate that the LamB1 IRES is activated by binding to La which leads to translational upregulation during hepatocellular EMT.
Collapse
Affiliation(s)
- Michaela Petz
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna and Institute for Animal Breeding and Genetics, University of Veterinary Medicine I, Veterinärplatz 1, 1210 Vienna, Austria
| | - Nicole Them
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna and Institute for Animal Breeding and Genetics, University of Veterinary Medicine I, Veterinärplatz 1, 1210 Vienna, Austria
| | - Heidemarie Huber
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna and Institute for Animal Breeding and Genetics, University of Veterinary Medicine I, Veterinärplatz 1, 1210 Vienna, Austria
| | - Hartmut Beug
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna and Institute for Animal Breeding and Genetics, University of Veterinary Medicine I, Veterinärplatz 1, 1210 Vienna, Austria
| | - Wolfgang Mikulits
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center Vienna, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna and Institute for Animal Breeding and Genetics, University of Veterinary Medicine I, Veterinärplatz 1, 1210 Vienna, Austria
| |
Collapse
|
63
|
Sommer G, Rossa C, Chi AC, Neville BW, Heise T. Implication of RNA-binding protein La in proliferation, migration and invasion of lymph node-metastasized hypopharyngeal SCC cells. PLoS One 2011; 6:e25402. [PMID: 22016766 PMCID: PMC3189910 DOI: 10.1371/journal.pone.0025402] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 09/02/2011] [Indexed: 01/22/2023] Open
Abstract
The 5-year survival rate for oral cavity cancer is poorer than for breast, colon or prostate cancer, and has improved only slightly in the last three decades. Hence, new therapeutic strategies are urgently needed. Here we demonstrate by tissue micro array analysis for the first time that RNA-binding protein La is significantly overexpressed in oral squamous cell carcinoma (SCC). Within this study we therefore addressed the question whether siRNA-mediated depletion of the La protein may interfere with known tumor-promoting characteristics of head and neck SCC cells. Our studies demonstrate that the La protein promotes cell proliferation, migration and invasion of lymph node-metastasized hypopharyngeal SCC cells. We also reveal that La is required for the expression of β-catenin as well as matrix metalloproteinase type 2 (MMP-2) within these cells. Taken together these data suggest a so far unknown function of the RNA-binding protein La in promoting tumor progression of head and neck SCC.
Collapse
Affiliation(s)
- Gunhild Sommer
- Department of Biochemistry and Molecular Biology, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America.
| | | | | | | | | |
Collapse
|
64
|
Requirement of rRNA methylation for 80S ribosome assembly on a cohort of cellular internal ribosome entry sites. Mol Cell Biol 2011; 31:4482-99. [PMID: 21930789 DOI: 10.1128/mcb.05804-11] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Protein syntheses mediated by cellular and viral internal ribosome entry sites (IRESs) are believed to have many features in common. Distinct mechanisms for ribosome recruitment and preinitiation complex assembly between the two processes have not been identified thus far. Here we show that the methylation status of rRNA differentially influenced the mechanism of 80S complex formation on IRES elements from the cellular sodium-coupled neutral amino acid transporter 2 (SNAT2) versus the hepatitis C virus mRNA. Translation initiation involves the assembly of the 48S preinitiation complex, followed by joining of the 60S ribosomal subunit and formation of the 80S complex. Abrogation of rRNA methylation did not affect the 48S complex but resulted in impairment of 80S complex assembly on the cellular, but not the viral, IRESs tested. Impairment of 80S complex assembly on the amino acid transporter SNAT2 IRES was rescued by purified 60S subunits containing fully methylated rRNA. We found that rRNA methylation did not affect the activity of any of the viral IRESs tested but affected the activity of numerous cellular IRESs. This work reveals a novel mechanism operating on a cohort of cellular IRESs that involves rRNA methylation for proper 80S complex assembly and efficient translation initiation.
Collapse
|
65
|
Stumpf CR, Ruggero D. The cancerous translation apparatus. Curr Opin Genet Dev 2011; 21:474-83. [PMID: 21543223 PMCID: PMC3481834 DOI: 10.1016/j.gde.2011.03.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 03/25/2011] [Indexed: 01/04/2023]
Abstract
Deregulations in translational control are critical features of cancer initiation and progression. Activation of key oncogenic pathways promotes rapid and dramatic translational reprogramming, not simply by increasing overall protein synthesis, but also by modulating specific mRNA networks that promote cellular transformation. Additionally, ribosomopathies caused by mutations in ribosome components alter translational regulation leading to specific pathological features, including cancer susceptibility. Exciting advances in our understanding of translational control in cancer have illuminated a striking specificity innate to the translational apparatus. Characterizing this specificity will provide novel insights into how cells normally utilize translational control to modulate gene expression, how it is deregulated in cancer, and how these processes can be targeted to develop new cancer therapies.
Collapse
Affiliation(s)
- Craig R. Stumpf
- School of Medicine and Department of Urology, Helen Diller Family, Comprehensive Cancer Center, University of California, San Francisco, Helen, Diller Family Cancer Research Building Room 386, 1450 3rd Street, San Francisco, CA 94158-3110
| | - Davide Ruggero
- School of Medicine and Department of Urology, Helen Diller Family, Comprehensive Cancer Center, University of California, San Francisco, Helen, Diller Family Cancer Research Building Room 386, 1450 3rd Street, San Francisco, CA 94158-3110
| |
Collapse
|
66
|
Nakamura-López Y, Villegas-Sepúlveda N, Sarmiento-Silva RE, Gómez B. Intrinsic apoptotic pathway is subverted in mouse macrophages persistently infected by RSV. Virus Res 2011; 158:98-107. [DOI: 10.1016/j.virusres.2011.03.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2010] [Revised: 03/16/2011] [Accepted: 03/17/2011] [Indexed: 11/29/2022]
|
67
|
Garlapati S, Saraiya AA, Wang CC. A La autoantigen homologue is required for the internal ribosome entry site mediated translation of giardiavirus. PLoS One 2011; 6:e18263. [PMID: 21479239 PMCID: PMC3066225 DOI: 10.1371/journal.pone.0018263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 02/28/2011] [Indexed: 12/11/2022] Open
Abstract
Translation of Giardiavirus (GLV) mRNA is initiated at an internal ribosome entry site (IRES) in the viral transcript. The IRES localizes to a downstream portion of 5′ untranslated region (UTR) and a part of the early downstream coding region of the transcript. Recent studies indicated that the IRES does not require a pre-initiation complex to initiate translation but may directly recruit the small ribosome subunit with the help of a number of trans-activating protein factors. A La autoantigen homologue in the viral host Giardia lamblia, GlLa, was proposed as one of the potential trans-activating factors based on its specific binding to GLV-IRES in vitro. In this study, we further elucidated the functional role of GlLa in GLV-IRES mediated translation in Giardia by knocking down GlLa with antisense morpholino oligo, which resulted in a reduction of GLV-IRES activity by 40%. An over-expression of GlLa in Giardia moderately stimulated GLV-IRES activity by 20%. A yeast inhibitory RNA (IRNA), known to bind mammalian and yeast La autoantigen and inhibit Poliovirus and Hepatitis C virus IRES activities in vitro and in vivo, was also found to bind to GlLa protein in vitro and inhibited GLV-IRES function in vivo. The C-terminal domain of La autoantigen interferes with the dimerization of La and inhibits its function. An over-expression of the C-terminal domain (200–348aa) of GlLa in Giardia showed a dominant-negative effect on GLV-IRES activity, suggesting a potential inhibition of GlLa dimerization. HA tagged GlLa protein was detected mainly in the cytoplasm of Giardia, thus supporting a primary role of GlLa in translation initiation in Giardiavirus.
Collapse
Affiliation(s)
- Srinivas Garlapati
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Ashesh A. Saraiya
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Ching C. Wang
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
68
|
Yeh SH, Yang WB, Gean PW, Hsu CY, Tseng JT, Su TP, Chang WC, Hung JJ. Translational and transcriptional control of Sp1 against ischaemia through a hydrogen peroxide-activated internal ribosomal entry site pathway. Nucleic Acids Res 2011; 39:5412-23. [PMID: 21441538 PMCID: PMC3141265 DOI: 10.1093/nar/gkr161] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The exact mechanism underlying increases in Sp1 and the physiological consequences thereafter remains unknown. In rat primary cortical neurons, oxygen-glucose deprivation (OGD) causes an increase in H2O2 as well as Sp1 in early ischaemia but apparently does not change mRNA level or Sp1 stability. We hereby identified a longer 5′-UTR in Sp1 mRNA that contains an internal ribosome entry site (IRES) that regulates rapid and efficient translation of existing mRNAs. By using polysomal fragmentation and bicistronic luciferase assays, we found that H2O2 activates IRES-dependent translation. Thus, H2O2 or tempol, a superoxide dismutase-mimetic, increases Sp1 levels in OGD-treated neurons. Further, early-expressed Sp1 binds to Sp1 promoter to cause a late rise in Sp1 in a feed-forward manner. Short hairpin RNA against Sp1 exacerbates OGD-induced apoptosis in primary neurons. While Sp1 levels increase in the cortex in a rat model of stroke, inhibition of Sp1 binding leads to enhanced apoptosis and cortical injury. These results demonstrate that neurons can use H2O2 as a signalling molecule to quickly induce Sp1 translation through an IRES-dependent translation pathway that, in cooperation with a late rise in Sp1 via feed-forward transcriptional activation, protects neurons against ischaemic damage.
Collapse
Affiliation(s)
- Shiu Hwa Yeh
- Department of Pharmacology, Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng-Kung University, Tainan 701, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Abstract
The majority of mRNAs in eukaryotic cells are translated via a method that is dependent upon the recognition of, and binding to, the methylguanosine cap at the 5' end of the mRNA, by a set of protein factors termed eIFs (eukaryotic initiation factors). However, many of the eIFs involved in this process are modified and become less active under a number of pathophysiological stress conditions, including amino acid starvation, heat shock, hypoxia and apoptosis. During these conditions, the continued synthesis of proteins essential to recovery from stress or maintenance of a cellular programme is mediated via an alternative form of translation initiation termed IRES (internal ribosome entry site)-mediated translation. This relies on the mRNA containing a complex cis-acting structural element in its 5'-UTR (untranslated region) that is able to recruit the ribosome independently of the cap, and is often dependent upon additional factors termed ITAFs (IRES trans-acting factors). A limited number of ITAFs have been identified to date, particularly for cellular IRESs, and it is not yet fully understood how they exert their control and which cellular pathways are involved in their regulation.
Collapse
|
70
|
Sommer G, Dittmann J, Kuehnert J, Reumann K, Schwartz PE, Will H, Coulter BL, Smith MT, Heise T. The RNA-binding protein La contributes to cell proliferation and CCND1 expression. Oncogene 2011; 30:434-44. [PMID: 20856207 DOI: 10.1038/onc.2010.425] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 07/22/2010] [Accepted: 08/10/2010] [Indexed: 11/08/2022]
Abstract
The La protein is an essential RNA-binding protein implicated in different aspects of RNA metabolism. Herein, we report that small interfering (siRNA)-mediated La depletion reduces cell proliferation of different cell lines concomitant with a reduction in cyclin D1 (CCND1) protein. To exclude off-target effects we demonstrate that exogenous La expression in La-depleted cells restores cell proliferation and CCND1 protein levels. In contrast, proliferation of immortalized CCND1 knockout cells is not affected by La depletion, supporting a functional coherence between La, CCND1 and proliferation. Furthermore, we document by reversible in vivo crosslinking and ribonucleoprotein (RNP) immunoprecipitation an association of the La protein with CCND1 messengerRNA and that CCND1 internal ribosome entry site (IRES)-dependent translation is modulated by La protein level within the cell. In addition, we show elevated La protein expression in cervical cancer tissue and its correlation with aberrant CCND1 protein levels in cervical tumor tissue lysates. In conclusion, this study establishes a role of La in cell proliferation and CCND1 expression and demonstrates for the first time an overexpression of the RNA-binding protein La in solid tumors.
Collapse
Affiliation(s)
- G Sommer
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
RNA-binding protein HuR mediates cytoprotection through stimulation of XIAP translation. Oncogene 2010; 30:1460-9. [PMID: 21102524 DOI: 10.1038/onc.2010.527] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Expression of the intrinsic cellular caspase inhibitor XIAP is regulated primarily at the level of protein synthesis. The 5' untranslated region harbours an Internal Ribosome Entry Site (IRES) motif that supports cap-independent translation of XIAP mRNA during conditions of cellular stress. In this study, we show that the RNA-binding protein HuR, which is known to orchestrate an antiapoptotic cellular program, stimulates translation of XIAP mRNA through XIAP IRES. We further show that HuR binds to XIAP IRES in vitro and in vivo, and stimulates recruitment of the XIAP mRNA into polysomes. Importantly, protection from the apoptosis-inducing agent etoposide by overexpression of HuR requires the presence of XIAP, suggesting that HuR-mediated cytoprotection is partially executed through enhanced XIAP translation. Our data suggest that XIAP belongs to the HuR-regulated RNA operon of antiapoptotic genes, which, along with Bcl-2, Mcl-1 and ProTα, contributes to the regulation of cell survival.
Collapse
|
72
|
Genolet R, Rahim G, Gubler-Jaquier P, Curran J. The translational response of the human mdm2 gene in HEK293T cells exposed to rapamycin: a role for the 5'-UTRs. Nucleic Acids Res 2010; 39:989-1003. [PMID: 20876686 PMCID: PMC3035446 DOI: 10.1093/nar/gkq805] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Polysomal messenger RNA (mRNA) populations change rapidly in response to alterations in the physiological status of the cell. For this reason, translational regulation, mediated principally at the level of initiation, plays a key role in the maintenance of cellular homeostasis. In an earlier translational profiling study, we followed the impact of rapamycin on polysome re-seeding. Despite the overall negative effect on transcript recruitment, we nonetheless observed that some mRNAs were significantly less affected. Consequently, their relative polysomal occupancy increased in the rapamycin-treated cells. The behaviour of one of these genes, mdm2, has been further analysed. Despite the absence of internal ribosome entry site activity we demonstrate, using a dual reporter assay, that both the reported mdm2 5′-UTRs confer resistance to rapamycin relative to the 5′-UTR of β-actin. This relative resistance is responsive to the downstream targets mTORC1 but did not respond to changes in the La protein, a reported factor acting positively on MDM2 translational expression. Furthermore, extended exposure to rapamycin in the presence of serum increased the steady-state level of the endogenous MDM2 protein. However, this response was effectively reversed when serum levels were reduced. Taken globally, these studies suggest that experimental conditions can dramatically modulate the expressional output during rapamycin exposure.
Collapse
Affiliation(s)
- Raphael Genolet
- Department of Microbiology and Molecular Medicine, University of Geneva Medical School (CMU) 1, rue Michel Servet, CH-1205 Geneva, Switzerland
| | | | | | | |
Collapse
|
73
|
Hundsdoerfer P, Dietrich I, Schmelz K, Eckert C, Henze G. XIAP expression is post-transcriptionally upregulated in childhood ALL and is associated with glucocorticoid response in T-cell ALL. Pediatr Blood Cancer 2010; 55:260-6. [PMID: 20582956 DOI: 10.1002/pbc.22541] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Resistance to glucocorticoid induced apoptosis is one of the major risk factors for relapse and poor outcome in childhood acute lymphoblastic leukemia (ALL). Overexpression of X-linked inhibitor of apoptosis protein (XIAP) has been shown to be associated with chemotherapy resistance in several malignancies. PROCEDURE XIAP protein and mRNA expression were determined in leukemic blasts of 51 childhood ALL patients and normal bone marrow mononuclear cells. XIAP expression was correlated with glucocorticoid response and outcome. RESULTS XIAP protein but not mRNA expression was found to be highly increased in childhood ALL compared to control bone marrow mononuclear cells (MNC) (median: 3.5 vs. 0.14 ng/10(5) MNC, P < 0.0001) indicating a post-transcriptional regulation of XIAP expression. In patients with T-cell ALL, poor prednisone response was associated with increased XIAP expression (median: 2.8 in good vs. 5.8 in poor responders; P = 0.005). Similarly, T-cell ALL patients suffering adverse events showed higher initial XIAP levels than patients in continuous complete remission (CCR) (median: 2.7 in patients in CCR vs. 5.6 in patients suffering adverse events; P = 0.007). XIAP inhibition using the low-molecular-weight SMAC mimetic LBW242 resulted in a significant increase of prednisone-induced apoptosis in vitro. CONCLUSION In childhood ALL compared to control bone marrow, the expression of the apoptosis inhibitor XIAP is highly increased by post-transcriptional regulation. The association with poor in vivo glucocorticoid response and outcome in T-cell ALL suggests XIAP inhibition as a promising novel approach for the treatment of resistant ALL.
Collapse
Affiliation(s)
- Patrick Hundsdoerfer
- Department of Pediatric Oncology/Hematology, Charité Universitätsmedizin, Berlin, Germany.
| | | | | | | | | |
Collapse
|
74
|
Röther S, Burkert C, Brünger KM, Mayer A, Kieser A, Strässer K. Nucleocytoplasmic shuttling of the La motif-containing protein Sro9 might link its nuclear and cytoplasmic functions. RNA (NEW YORK, N.Y.) 2010; 16:1393-1401. [PMID: 20494970 PMCID: PMC2885688 DOI: 10.1261/rna.2089110] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 04/01/2010] [Indexed: 05/29/2023]
Abstract
Diverse steps in gene expression are tightly coupled. Curiously, the La-motif-containing protein Sro9 has been shown to play a role in transcription and translation. Here, we show that Sro9 interacts with nuclear and cytoplasmic protein complexes involved in gene expression. In addition, Sro9 shuttles between nucleus and cytoplasm and is exported from the nucleus in an mRNA export-dependent manner. Importantly, Sro9 is recruited to transcribed genes. However, whole genome expression analysis shows that loss of Sro9 function does not greatly change the level of specific transcripts indicating that Sro9 does not markedly affect their synthesis and/or stability. Taken together, Sro9 might bind to the mRNP already during transcription and accompany the mature mRNP to the cytoplasm where it modulates translation of the mRNA.
Collapse
Affiliation(s)
- Susanne Röther
- Department of Biochemistry, Ludwig-Maximilians-University Munich, Gene Center and Center for Integrated Protein Science Munich, 81377 Munich, Germany
| | | | | | | | | | | |
Collapse
|
75
|
Mishra PJ, Menon LG, Mishra PJ, Mayer-Kuckuk P, Bertino JR, Banerjee D. Translational modulation of proteins expressed from bicistronic vectors. Mol Imaging 2009; 8:305-18. [PMID: 20003889 PMCID: PMC2864087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023] Open
Abstract
Bicistronic vectors are useful tools for exogenous expression of two gene products from a single promoter element; however, reduced expression of protein from the second cistron compared with the first cistron is a common limitation to this approach. To overcome this limitation, we explored use of dihydrofolate reductase (DHFR) complementary DNA encoded in bicistronic vectors to induce a second protein of interest by methotrexate (MTX) treatment. Previous studies have demonstrated that levels of DHFR protein and DHFR fusion protein can be induced translationally following MTX treatment of cells. We demonstrated that in response to MTX treatment, DHFR partner protein in a bicistronic construct is induced for longer periods of time when compared with endogenous DHFR and DHFR fusion protein, in vitro and in vivo. Using rapamycin pretreatment followed by MTX treatment, we also devised a strategy to modulate levels of two proteins expressed from a bicistronic construct in a cap-independent manner. To our knowledge, this is the first report demonstrating that levels of proteins in DHFR-based bicistronic constructs can be induced and modulated using MTX and rapamycin treatment.
Collapse
Affiliation(s)
- Prasun J Mishra
- Department of Pharmacology, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, USA
| | | | | | | | | | | |
Collapse
|
76
|
Graber TE, Baird SD, Kao PN, Mathews MB, Holcik M. NF45 functions as an IRES trans-acting factor that is required for translation of cIAP1 during the unfolded protein response. Cell Death Differ 2009; 17:719-29. [PMID: 19893574 DOI: 10.1038/cdd.2009.164] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Expression of the cellular inhibitor of apoptosis protein 1 (cIAP1) is unexpectedly repressed at the level of translation under normal physiological conditions in many cell lines. We have previously shown that the 5' untranslated region of cIAP1 mRNA contains a stress-inducible internal ribosome entry site (IRES) that governs expression of cIAP1 protein. Although inactive in unstressed cells, the IRES supports cap-independent translation of cIAP1 in response to endoplasmic reticulum stress. To gain an insight into the mechanism of cIAP1 IRES function, we empirically derived the minimal free energy secondary structure of the cIAP1 IRES using enzymatic cleavage mapping. We subsequently used RNA affinity chromatography to identify several cellular proteins, including nuclear factor 45 (NF45) as cIAP1 IRES binding proteins. In this report we show that NF45 is a novel RNA binding protein that enhances IRES-dependent translation of endogenous cIAP1. Further, we show that NF45 is required for IRES-mediated induction of cIAP1 protein during the unfolded protein response. The data presented are consistent with a model in which translation of cIAP1 is governed, at least in part, by NF45, a novel cellular IRES trans-acting factor.
Collapse
Affiliation(s)
- T E Graber
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | | | | | | | | |
Collapse
|
77
|
Mishra PJ, Menon LG, Mishra PJ, Mayer-Kuckuk P, Bertino JR, Banerjee D. Translational Modulation of Proteins Expressed from Bicistronic Vectors. Mol Imaging 2009. [DOI: 10.2310/7290.2009.00028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Prasun J. Mishra
- From the Department of Pharmacology, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Department of Neurosurgery, Brigham and Women's
| | - Lata G. Menon
- From the Department of Pharmacology, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Department of Neurosurgery, Brigham and Women's
| | - Pravin J. Mishra
- From the Department of Pharmacology, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Department of Neurosurgery, Brigham and Women's
| | - Philipp Mayer-Kuckuk
- From the Department of Pharmacology, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Department of Neurosurgery, Brigham and Women's
| | - Joseph R. Bertino
- From the Department of Pharmacology, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Department of Neurosurgery, Brigham and Women's
| | - Debabrata Banerjee
- From the Department of Pharmacology, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD; Department of Medicine, Robert Wood Johnson Medical School, Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Department of Neurosurgery, Brigham and Women's
| |
Collapse
|
78
|
Dhar D, Venkataramana M, Ponnuswamy A, Das S. Role of polypyrimidine tract binding protein in mediating internal initiation of translation of interferon regulatory factor 2 RNA. PLoS One 2009; 4:e7049. [PMID: 19756143 PMCID: PMC2737629 DOI: 10.1371/journal.pone.0007049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 07/14/2009] [Indexed: 11/19/2022] Open
Abstract
Background Earlier we have reported translational control of interferon regulatory factor 2 (IRF2) by internal initiation (Dhar et al, Nucleic Acids Res, 2007). The results implied possible role of IRF2 in controlling the intricate balance of cellular gene expression under stress conditions in general. Here we have investigated the secondary structure of the Internal Ribosome Entry Site of IRF2 RNA and demonstrated the role of PTB protein in ribosome assembly to facilitate internal initiation. Methodology/Principal Findings We have probed the putative secondary structure of the IRF2 5′UTR RNA using various enzymatic and chemical modification agents to constrain the secondary structure predicted from RNA folding algorithm Mfold. The IRES activity was found to be influenced by the interaction of trans-acting factor, polypyrimidine tract binding protein (PTB). Deletion of 25 nts from the 3′terminus of the 5′untranslated region resulted in reduced binding with PTB protein and also showed significant decrease in IRES activity compared to the wild type. We have also demonstrated putative contact points of PTB on the IRF2–5′UTR using primer extension inhibition assay. Majority of the PTB toe-prints were found to be restricted to the 3′end of the IRES. Additionally, Circular Dichroism (CD) spectra analysis suggested change in the conformation of the RNA upon PTB binding. Further, binding studies using S10 extract from HeLa cells, partially silenced for PTB gene expression, resulted in reduced binding by other trans-acting factors. Finally, we have demonstrated that addition of recombinant PTB enhances ribosome assembly on IRF2 IRES suggesting possible role of PTB in mediating internal initiation of translation of IRF2 RNA. Conclusion/Significance It appears that PTB binding to multiple sites within IRF2 5′UTR leads to a conformational change in the RNA that facilitate binding of other trans-acting factors to mediate internal initiation of translation.
Collapse
Affiliation(s)
- Debojyoti Dhar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Musturi Venkataramana
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Anand Ponnuswamy
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Saumitra Das
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
- * E-mail:
| |
Collapse
|
79
|
Gu L, Zhu N, Zhang H, Durden DL, Feng Y, Zhou M. Regulation of XIAP translation and induction by MDM2 following irradiation. Cancer Cell 2009; 15:363-75. [PMID: 19411066 PMCID: PMC2696306 DOI: 10.1016/j.ccr.2009.03.002] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 12/23/2008] [Accepted: 03/02/2009] [Indexed: 02/02/2023]
Abstract
Increases in protein levels of XIAP in cancer cells have been associated with resistance to apoptosis induced by cellular stress. Herein we demonstrate that the upregulation of XIAP protein levels is regulated by MDM2 at the translational level. MDM2 was found to physically interact with the IRES of the XIAP 5'-UTR, and to positively regulate XIAP IRES activity. This XIAP IRES-dependent translation was significantly increased in MDM2-transfected cells where MDM2 accumulated in the cytoplasm. Cellular stress and DNA damage triggered by irradiation induced the dephosphorylation and cytoplasmic localization of MDM2, which also led to an increase in IRES-dependent XIAP translation. Upregulation of XIAP in MDM2-overexpressing cancer cells in response to irradiation resulted in resistance of these cells to radiation-induced apoptosis.
Collapse
Affiliation(s)
- Lubing Gu
- Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Emory University School of Medicine, Atlanta, GA 30322
| | - Ningxi Zhu
- Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Emory University School of Medicine, Atlanta, GA 30322
| | - Hongying Zhang
- Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Emory University School of Medicine, Atlanta, GA 30322
| | - Donald L. Durden
- Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Emory University School of Medicine, Atlanta, GA 30322
| | - Yue Feng
- Department of Pharmacology, Aflac Cancer Center and Blood Disorders Service, Emory University School of Medicine, Atlanta, GA 30322
| | - Muxiang Zhou
- Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
80
|
Bousquet-Antonelli C, Deragon JM. A comprehensive analysis of the La-motif protein superfamily. RNA (NEW YORK, N.Y.) 2009; 15:750-64. [PMID: 19299548 PMCID: PMC2673062 DOI: 10.1261/rna.1478709] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 01/22/2009] [Indexed: 05/24/2023]
Abstract
The extremely well-conserved La motif (LAM), in synergy with the immediately following RNA recognition motif (RRM), allows direct binding of the (genuine) La autoantigen to RNA polymerase III primary transcripts. This motif is not only found on La homologs, but also on La-related proteins (LARPs) of unrelated function. LARPs are widely found amongst eukaryotes and, although poorly characterized, appear to be RNA-binding proteins fulfilling crucial cellular functions. We searched the fully sequenced genomes of 83 eukaryotic species scattered along the tree of life for the presence of LAM-containing proteins. We observed that these proteins are absent from archaea and present in all eukaryotes (except protists from the Plasmodium genus), strongly suggesting that the LAM is an ancestral motif that emerged early after the archaea-eukarya radiation. A complete evolutionary and structural analysis of these proteins resulted in their classification into five families: the genuine La homologs and four LARP families. Unexpectedly, in each family a conserved domain representing either a classical RRM or an RRM-like motif immediately follows the LAM of most proteins. An evolutionary analysis of the LAM-RRM/RRM-L regions shows that these motifs co-evolved and should be used as a single entity to define the functional region of interaction of LARPs with their substrates. We also found two extremely well conserved motifs, named LSA and DM15, shared by LARP6 and LARP1 family members, respectively. We suggest that members of the same family are functional homologs and/or share a common molecular mode of action on different RNA baits.
Collapse
|
81
|
Carter JR, Fraser TS, Fraser MJ. Examining the relative activity of several dicistrovirus intergenic internal ribosome entry site elements in uninfected insect and mammalian cell lines. J Gen Virol 2009; 89:3150-3155. [PMID: 19008405 DOI: 10.1099/vir.0.2008/003921-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Comparisons of the relative activities of 11 intergenic region (IGR) internal ribosome entry site (IRES) elements of insect dicistrovirus with 5' IRES elements of the hepatitis C and encephalomyocarditis viruses were performed in insect and mammalian cells. Dual luciferase assays were performed to determine the most effective dicistrovirus IGR IRES in the lepidopteran cell lines Sf9 (Spodoptera frugiperda) and BmN (Bombyx mori), and the dipteran cell lines S2 (Drosophila melanogaster) and ATC-10 (Aedes aegypti). Evaluation of dual luciferase expression from DNA plasmids and in vitro-transcribed RNA revealed apparent splicing with certain IRES elements. Though IRES activity depended upon the cell line examined, the black queen cell and Drosophila C dicistrovirus intergenic IRES elements were most effective for coupled gene expression in the diverse insect cell lines examined.
Collapse
Affiliation(s)
- James R Carter
- 214 Galvin Life Sciences, Department of Biological Sciences, Center for Tropical Diseases Research and Training, University of Notre Dame, PO Box 369, Notre Dame, IN 46556-0369, USA
| | - Tresa S Fraser
- 214 Galvin Life Sciences, Department of Biological Sciences, Center for Tropical Diseases Research and Training, University of Notre Dame, PO Box 369, Notre Dame, IN 46556-0369, USA
| | - Malcolm J Fraser
- 214 Galvin Life Sciences, Department of Biological Sciences, Center for Tropical Diseases Research and Training, University of Notre Dame, PO Box 369, Notre Dame, IN 46556-0369, USA
| |
Collapse
|
82
|
Cecconi D, Zamò A, Bianchi E, Parisi A, Barbi S, Milli A, Rinalducci S, Rosenwald A, Hartmann E, Zolla L, Chilosi M. Signal transduction pathways of mantle cell lymphoma: A phosphoproteome-based study. Proteomics 2008; 8:4495-506. [DOI: 10.1002/pmic.200800080] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
83
|
Kazadi K, Loeuillet C, Deutsch S, Ciuffi A, Muñoz M, Beckmann JS, Moradpour D, Antonarakis SE, Telenti A. Genomic determinants of the efficiency of internal ribosomal entry sites of viral and cellular origin. Nucleic Acids Res 2008; 36:6918-25. [PMID: 18978018 PMCID: PMC2588522 DOI: 10.1093/nar/gkn812] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Variation in cellular gene expression levels has been shown to be inherited. Expression is controlled at transcriptional and post-transcriptional levels. Internal ribosome entry sites (IRES) are used by viruses to bypass inhibition of cap-dependent translation, and by eukaryotic cells to control translation under conditions when protein synthesis is inhibited. We aimed at identifying genomic determinants of variability in IRES-mediated translation of viral [Encephalomyocarditis virus (EMCV)] and cellular IRES [X-linked inhibitor-of-apoptosis (XIAP) and c-myc]. Bicistronic lentiviral constructs expressing two fluorescent reporters were used to transduce laboratory and B lymphoblastoid cell lines [15 CEPH pedigrees (n = 205) and 50 unrelated individuals]. IRES efficiency varied according to cell type and among individuals. Control of IRES activity has a significant genetic component (h2 of 0.47 and 0.36 for EMCV and XIAP, respectively). Quantitative linkage analysis identified a suggestive locus (LOD 2.35) on chromosome 18q21.2, and genome-wide association analysis revealed of a cluster of SNPs on chromosome 3, intronic to the FHIT gene, marginally associated (P = 5.9E-7) with XIAP IRES function. This study illustrates the in vitro generation of intermediate phenotypes by using cell lines for the evaluation of genetic determinants of control of elements such as IRES.
Collapse
Affiliation(s)
- Kayole Kazadi
- Institute of Microbiology, University Hospital Center, University of Lausanne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Contribution of internal initiation to translation of cellular mRNAs containing IRESs. Biochem Soc Trans 2008; 36:694-7. [DOI: 10.1042/bst0360694] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A broad range of cellular stresses lead to the inhibition of translation. Despite this, some cellular mRNAs are selectively translated under these conditions. It is widely supposed that cap-independent internal initiation may maintain efficient translation of particular cellular mRNAs under a variety of stresses and other special conditions when cap-dependent protein synthesis is impaired. However, in spite of a large number of reports focused on the investigation of the regulation of IRES (internal ribosome entry site) activity in different tissues and under various stresses, only rarely is the real efficiency of IRES-driven translation in comparison with cap-dependent translation evaluated. When precisely measured, the efficiencies of candidate IRESs in most cases appeared to be very low and not sufficient to compensate for the reduction of cap-dependent initiation under stresses. The usually low efficiency of internal initiation of translation is inconsistent with postulated biological roles of IRESs.
Collapse
|
85
|
Kotik-Kogan O, Valentine ER, Sanfelice D, Conte MR, Curry S. Structural analysis reveals conformational plasticity in the recognition of RNA 3' ends by the human La protein. Structure 2008; 16:852-62. [PMID: 18547518 PMCID: PMC2430598 DOI: 10.1016/j.str.2008.02.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 02/19/2008] [Accepted: 02/24/2008] [Indexed: 01/29/2023]
Abstract
The eukaryotic La protein recognizes the 3' poly(U) sequences of nascent RNA polymerase III transcripts to assist folding and maturation. The 3' ends of such RNAs are bound by the N-terminal domain of La (LaNTD). We have solved the crystal structures of four LaNTD:RNA complexes, each containing a different single-stranded RNA oligomer, and compared them to the structure of a previously published LaNTD:RNA complex containing partially duplex RNA. The presence of purely single-stranded RNA in the binding pocket at the interface between the La motif and RRM domains allows significantly closer contact with the 3' end of the RNA. Comparison of the different LaNTD:RNA complexes identifies a conserved set of interactions with the last two nucleotides at the 3' end of the RNA ligand that are key to binding. Strikingly, we also observe two alternative conformations of bound ssRNA, indicative of an unexpected degree of plasticity in the modes of RNA binding.
Collapse
Affiliation(s)
- Olga Kotik-Kogan
- Biophysics Section, Blackett Laboratory, Imperial College, Exhibition Road, London SW7 2AZ, United Kingdom
| | - Elizabeth R. Valentine
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, United Kingdom
| | - Domenico Sanfelice
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, United Kingdom
| | - Maria R. Conte
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, United Kingdom
| | - Stephen Curry
- Biophysics Section, Blackett Laboratory, Imperial College, Exhibition Road, London SW7 2AZ, United Kingdom
| |
Collapse
|
86
|
Concomitant transitory up-regulation of X-linked inhibitor of apoptosis protein (XIAP) and the heterogeneous nuclear ribonucleoprotein C1-C2 in surviving cells during neuronal apoptosis. Neurochem Res 2008; 33:1859-68. [PMID: 18363099 DOI: 10.1007/s11064-008-9658-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Accepted: 03/07/2008] [Indexed: 10/22/2022]
Abstract
Although cap-dependent translation initiation is the prevalent mode of ribosome binding to mRNAs in eukaryotes, some mRNAs exhibit the ability to bypass the requirement for the cap structure. The translation of X-chromosome-linked inhibitor of apoptosis protein (XIAP) mRNA is controlled by an internal ribosome entry site (IRES) element, which requires the interaction of the heterogeneous nuclear ribonucleoprotein C1-C2 (hnRNP-C1/C2). We analyze, at the protein level, the time course and distribution of XIAP and hnRNP-C1/C2 upon ischemia in mice or staurosporine (STP)-induced apoptosis in HT22 cells. Both ischemia and STP induced a parallel upregulation of XIAP and hnRNP-C1/C2 protein levels in the penumbra and in HT22 cells. These results suggest that the increased levels of hnRNP C1/C2 may modulate XIAP translation, probably by interacting with the XIAP-IRES. The up-regulation of hnRNP-C1/C2 may foster the synthesis of XIAP as a protective pathway by which neurons try to counteract the initial deleterious effects of apoptosis.
Collapse
|
87
|
Beck ET, Blair CD, Black WC, Beaty BJ, Blitvich BJ. Alternative splicing generates multiple transcripts of the inhibitor of apoptosis protein 1 in Aedes and Culex spp. mosquitoes. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2007; 37:1222-1233. [PMID: 17916508 PMCID: PMC2065863 DOI: 10.1016/j.ibmb.2007.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 07/16/2007] [Accepted: 07/20/2007] [Indexed: 05/25/2023]
Abstract
We determined the sequences of cDNA encoding Inhibitor of Apoptosis Protein 1 (IAP1) homologues from Aedes triseriatus, Aedes albopictus, Aedes aegypti, Culex pipiens and Culex tarsalis. The cDNAs encode translation products that share > or = 84% sequence similarity. The IAP1 mRNA of each mosquito species exists as 3-5 distinct variants due to the presence of heterogeneous sequences at the distal end of their 5'UTRs. Partial genomic sequencing upstream of the 5' end of the Ae. triseriatus IAP1 gene, and analysis of the Ae. aegypti genomic sequence, suggest that these mRNA variants are generated by alternative splicing. Each IAP1 mRNA variant from Ae. triseriatus and Cx. pipiens was detected by RT-PCR in all mosquito life-stages and adult tissues examined, and the relative concentration of each Ae. triseriatus IAP mRNA variant in various tissues was determined.
Collapse
Affiliation(s)
- Eric T Beck
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523-1692, USA
| | | | | | | | | |
Collapse
|
88
|
Abstract
Apoptosis has been accepted as a fundamental component in the pathogenesis of cancer, in addition to other human diseases including neurodegeneration, coronary disease and diabetes. The origin of cancer involves deregulated cellular proliferation and the suppression of apoptotic processes, ultimately leading to tumor establishment and growth. Several lines of evidence point toward the IAP family of proteins playing a role in oncogenesis, via their effective suppression of apoptosis. The central mechanisms of IAP apoptotic suppression appear to be through direct caspase and pro-caspase inhibition (primarily caspase 3 and 7) and modulation of, and by, the transcription factor NF-kappaB. Thus, when the IAPs are over-expressed or over-active, as is the case in many cancers, cells are no longer able to die in a physiologically programmed fashion and become increasingly resistant to standard chemo- and radiation therapies. To date several approaches have been taken to target and eliminate IAP function in an attempt to re-establish sensitivity, reduce toxicity, and improve efficacy of cancer treatment. In this review, we address IAP proteins as therapeutic targets for the treatment of cancer and emphasize the importance of novel therapeutic approaches for cancer therapy. Novel targets of IAP function are being identified and include gene therapy strategies and small molecule inhibitors that are based on endogenous IAP antagonists. As well, molecular mechanistic approaches, such as RNAi to deplete IAP expression, are in development.
Collapse
Affiliation(s)
- Allison M Hunter
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, ON, Canada, K1H 8L1
| | | | | |
Collapse
|
89
|
Timmerman SL, Pfingsten JS, Kieft JS, Krushel LA. The 5' leader of the mRNA encoding the mouse neurotrophin receptor TrkB contains two internal ribosomal entry sites that are differentially regulated. PLoS One 2007; 3:e3242. [PMID: 18779873 PMCID: PMC2531235 DOI: 10.1371/journal.pone.0003242] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Accepted: 08/27/2008] [Indexed: 12/30/2022] Open
Abstract
A single internal ribosomal entry site (IRES) in conjunction with IRES transactivating factors (ITAFs) is sufficient to recruit the translational machinery to a eukaryotic mRNA independent of the cap structure. However, we demonstrate that the mouse TrkB mRNA contains two independent IRESes. The mouse TrkB mRNA consists of one of two 5′ leaders (1428 nt and 448 nt), both of which include the common 3′ exon (Ex2, 344 nt). Dicistronic RNA transfections and in vitro translation of monocistronic RNA demonstrated that both full-length 5′ leaders, as well as Ex2, exhibit IRES activity indicating the IRES is located within Ex2. Additional analysis of the upstream sequences demonstrated that the first 260 nt of exon 1 (Ex1a) also contains an IRES. Dicistronic RNA transfections into SH-SY5Y cells showed the Ex1a IRES is constitutively active. However, the Ex2 IRES is only active in response to retinoic acid induced neural differentiation, a state which correlates with the synthesis of the ITAF polypyrimidine tract binding protein (PTB1). Correspondingly, addition or knock-down of PTB1 altered Ex2, but not Ex1a IRES activity in vitro and ex vivo, respectively. These results demonstrate that the two functionally independent IRESes within the mouse TrkB 5′ leader are differentially regulated, in part by PTB1.
Collapse
Affiliation(s)
- Stephanie L. Timmerman
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
| | - Jennifer S. Pfingsten
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
| | - Jeffrey S. Kieft
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
| | - Les A. Krushel
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
- Department of Pharmacology, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
90
|
Lin JH, Dutz JP, Sontheimer RD, Werth VP. Pathophysiology of Cutaneous Lupus Erythematosus. Clin Rev Allergy Immunol 2007; 33:85-106. [DOI: 10.1007/s12016-007-0031-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
91
|
Tun C, Guo W, Nguyen H, Yun B, Libby RT, Morrison RS, Garden GA. Activation of the extrinsic caspase pathway in cultured cortical neurons requires p53-mediated down-regulation of the X-linked inhibitor of apoptosis protein to induce apoptosis. J Neurochem 2007; 102:1206-19. [PMID: 17488272 DOI: 10.1111/j.1471-4159.2007.04609.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cultured cortical neurons exposed to the Human Immunodeficiency Virus gp120 coat protein undergo apoptosis involving activation of both caspase-8 and caspase-9. Additionally, gp120-mediated neuronal apoptosis requires the pro-apoptotic transcription factor p53. As caspase-8-induced apoptosis does not typically require p53, we examined the possibility of a novel role for p53 in caspase-8 activation initiated by gp120. We observed that gp120 treatment of cultured cortical neurons induced caspase-8 activity and Bid cleavage independently of p53, but induction of caspase-3 enzymatic activity required p53 expression. These findings suggested the possibility that p53 down-regulates a caspase-3 inhibitor. We observed high-level expression of the caspase-3/9 inhibitor X-linked inhibitor of apoptosis protein (XIAP) in cultured cortical neurons. Adenoviral expression of p53 or induction of endogenous p53 by camptothecin treatment reduced XIAP protein in neurons. Infection with a p53 expressing adenovirus increased expression of the mRNA for Omi/HtrA2, a protease that cleaves and inactivates XIAP. These findings suggest that p53 regulates neuronal apoptosis, in part, by suppressing the anti-apoptotic protein XIAP via transcriptional activation of Omi/HtrA2.
Collapse
Affiliation(s)
- Christina Tun
- Department of Neurology, The University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | |
Collapse
|
92
|
van Niekerk EA, Willis DE, Chang JH, Reumann K, Heise T, Twiss JL. Sumoylation in axons triggers retrograde transport of the RNA-binding protein La. Proc Natl Acad Sci U S A 2007; 104:12913-8. [PMID: 17646655 PMCID: PMC1937566 DOI: 10.1073/pnas.0611562104] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A surprisingly large population of mRNAs has been shown to localize to sensory axons, but few RNA-binding proteins have been detected in these axons. These axonal mRNAs include several potential binding targets for the La RNA chaperone protein. La is transported into axonal processes in both culture and peripheral nerve. Interestingly, La is posttranslationally modified in sensory neurons by sumoylation. In axons, small ubiquitin-like modifying polypeptides (SUMO)-La interacts with dynein, whereas native La interacts with kinesin. Lysine 41 is required for sumoylation, and sumoylation-incompetent La(K41R) shows only anterograde transport, whereas WT La shows both anterograde and retrograde transport in axons. Thus, sumoylation of La determines the directionality of its transport within the axonal compartment, with SUMO-La likely recycling to the cell body.
Collapse
Affiliation(s)
- Erna A. van Niekerk
- *Department of Biological Sciences, University of Delaware, Newark, DE 19713
| | - Dianna E. Willis
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803
| | - Jay H. Chang
- Neural Development and Plasticity Section, Laboratory of Cellular and Synaptic Neurophysiology, National Institute of Child Health and Human Development–National Institutes of Health, Bethesda, MD 20892
| | - Kerstin Reumann
- Heinrich Pette Institute for Experimental Virology and Immunology, University of Hamburg, D-20251 Hamburg, Germany; and
| | - Tilman Heise
- Heinrich Pette Institute for Experimental Virology and Immunology, University of Hamburg, D-20251 Hamburg, Germany; and
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - Jeffery L. Twiss
- *Department of Biological Sciences, University of Delaware, Newark, DE 19713
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
93
|
Abstract
The ribosome filter hypothesis postulates that ribosomes are not simply translation machines but also function as regulatory elements that differentially affect or filter the translation of particular mRNAs. On the basis of new information, we take the opportunity here to review the ribosome filter hypothesis, suggest specific mechanisms of action, and discuss recent examples from the literature that support it.
Collapse
|
94
|
Baird SD, Lewis SM, Turcotte M, Holcik M. A search for structurally similar cellular internal ribosome entry sites. Nucleic Acids Res 2007; 35:4664-77. [PMID: 17591613 PMCID: PMC1950536 DOI: 10.1093/nar/gkm483] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 05/31/2007] [Accepted: 06/04/2007] [Indexed: 01/01/2023] Open
Abstract
Internal ribosome entry sites (IRES) allow ribosomes to be recruited to mRNA in a cap-independent manner. Some viruses that impair cap-dependent translation initiation utilize IRES to ensure that the viral RNA will efficiently compete for the translation machinery. IRES are also employed for the translation of a subset of cellular messages during conditions that inhibit cap-dependent translation initiation. IRES from viruses like Hepatitis C and Classical Swine Fever virus share a similar structure/function without sharing primary sequence similarity. Of the cellular IRES structures derived so far, none were shown to share an overall structural similarity. Therefore, we undertook a genome-wide search of human 5'UTRs (untranslated regions) with an empirically derived structure of the IRES from the key inhibitor of apoptosis, X-linked inhibitor of apoptosis protein (XIAP), to identify novel IRES that share structure/function similarity. Three of the top matches identified by this search that exhibit IRES activity are the 5'UTRs of Aquaporin 4, ELG1 and NF-kappaB repressing factor (NRF). The structures of AQP4 and ELG1 IRES have limited similarity to the XIAP IRES; however, they share trans-acting factors that bind the XIAP IRES. We therefore propose that cellular IRES are not defined by overall structure, as viral IRES, but are instead dependent upon short motifs and trans-acting factors for their function.
Collapse
Affiliation(s)
- Stephen D. Baird
- Department of Biochemistry, Microbiology and Immunology, Department of Pediatrics and School of Information Technology and Engineering, University of Ottawa, ON, Canada and Apoptosis Research Centre, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada, K1H 8L1
| | - Stephen M. Lewis
- Department of Biochemistry, Microbiology and Immunology, Department of Pediatrics and School of Information Technology and Engineering, University of Ottawa, ON, Canada and Apoptosis Research Centre, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada, K1H 8L1
| | - Marcel Turcotte
- Department of Biochemistry, Microbiology and Immunology, Department of Pediatrics and School of Information Technology and Engineering, University of Ottawa, ON, Canada and Apoptosis Research Centre, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada, K1H 8L1
| | - Martin Holcik
- Department of Biochemistry, Microbiology and Immunology, Department of Pediatrics and School of Information Technology and Engineering, University of Ottawa, ON, Canada and Apoptosis Research Centre, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada, K1H 8L1
| |
Collapse
|
95
|
Valavanis C, Wang Z, Sun D, Vaine M, Schwartz LM. Acheron, a novel member of the Lupus Antigen family, is induced during the programmed cell death of skeletal muscles in the moth Manduca sexta. Gene 2007; 393:101-9. [PMID: 17383118 PMCID: PMC2739619 DOI: 10.1016/j.gene.2007.01.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2006] [Revised: 01/24/2007] [Accepted: 01/26/2007] [Indexed: 11/15/2022]
Abstract
In order to identify novel genes associated with the initiation of programmed cell death during development, we employed a differential screening protocol to isolate cDNAs that were induced when the intersegmental muscles (ISM) of the moth Manduca sexta become committed to die at the end of metamorphosis. In this report we provide the first description of Acheron (Achn), a novel protein that was isolated in this screen. Acheron contains three Lupus antigen (La) repeats, nuclear localization and export (NLS and NES) signals, and an RNA recognition motif. Achn defines a new subfamily of La proteins that appears to have branched from authentic La protein relatively late in metazoan evolution. Achn is widely expressed in various insect, mouse and human tissues. Consistent with its expression during ISM death, Achn has been shown in separate studies to control muscle differentiation and apoptosis in both mice and zebrafish. These data define Achn as a newly discovered regulatory molecule that presumably mediates a variety of developmental and homeostatic processes in animals.
Collapse
Affiliation(s)
- Christos Valavanis
- Department of Biology, Morrill Science Center, University of Massachusetts, Amherst, Massachusetts 01003
| | - Zhaohui Wang
- Molecular and Cellular Biology Program, Morrill Science Center, University of Massachusetts, Amherst, Massachusetts 01003
| | - Danhui Sun
- Molecular and Cellular Biology Program, Morrill Science Center, University of Massachusetts, Amherst, Massachusetts 01003
| | - Michael Vaine
- Department of Biology, Morrill Science Center, University of Massachusetts, Amherst, Massachusetts 01003
| | - Lawrence M. Schwartz
- Department of Biology, Morrill Science Center, University of Massachusetts, Amherst, Massachusetts 01003
- Molecular and Cellular Biology Program, Morrill Science Center, University of Massachusetts, Amherst, Massachusetts 01003
- Pioneer Valley Life Sciences Institute, 3601 Main Street, Springfield, Massachusetts, 01199
| |
Collapse
|
96
|
Fleurdépine S, Deragon JM, Devic M, Guilleminot J, Bousquet-Antonelli C. A bona fide La protein is required for embryogenesis in Arabidopsis thaliana. Nucleic Acids Res 2007; 35:3306-21. [PMID: 17459889 PMCID: PMC1904278 DOI: 10.1093/nar/gkm200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 03/21/2007] [Accepted: 03/21/2007] [Indexed: 01/28/2023] Open
Abstract
Searches in the Arabidopsis thaliana genome using the La motif as query revealed the presence of eight La or La-like proteins. Using structural and phylogenetic criteria, we identified two putative genuine La proteins (At32 and At79) and showed that both are expressed throughout plant development but at different levels and under different regulatory conditions. At32, but not At79, restores Saccharomyces cerevisiae La nuclear functions in non-coding RNAs biogenesis and is able to bind to plant 3'-UUU-OH RNAs. We conclude that these La nuclear functions are conserved in Arabidopsis and supported by At32, which we renamed as AtLa1. Consistently, AtLa1 is predominantly localized to the plant nucleoplasm and was also detected in the nucleolar cavity. The inactivation of AtLa1 in Arabidopsis leads to an embryonic-lethal phenotype with deficient embryos arrested at early globular stage of development. In addition, mutant embryonic cells display a nucleolar hypertrophy suggesting that AtLa1 is required for normal ribosome biogenesis. The identification of two distantly related proteins with all structural characteristics of genuine La proteins suggests that these factors evolved to a certain level of specialization in plants. This unprecedented situation provides a unique opportunity to dissect the very different aspects of this crucial cellular activity.
Collapse
Affiliation(s)
- Sophie Fleurdépine
- CNRS UMR6547 GEEM, Université Blaise Pascal, 63177 Aubière, France and CNRS UMR5096 LGDP, Université de Perpignan Via Domitia, 66860 Perpignan, France
| | - Jean-Marc Deragon
- CNRS UMR6547 GEEM, Université Blaise Pascal, 63177 Aubière, France and CNRS UMR5096 LGDP, Université de Perpignan Via Domitia, 66860 Perpignan, France
| | - Martine Devic
- CNRS UMR6547 GEEM, Université Blaise Pascal, 63177 Aubière, France and CNRS UMR5096 LGDP, Université de Perpignan Via Domitia, 66860 Perpignan, France
| | - Jocelyne Guilleminot
- CNRS UMR6547 GEEM, Université Blaise Pascal, 63177 Aubière, France and CNRS UMR5096 LGDP, Université de Perpignan Via Domitia, 66860 Perpignan, France
| | - Cécile Bousquet-Antonelli
- CNRS UMR6547 GEEM, Université Blaise Pascal, 63177 Aubière, France and CNRS UMR5096 LGDP, Université de Perpignan Via Domitia, 66860 Perpignan, France
| |
Collapse
|
97
|
Kamrud KI, Custer M, Dudek JM, Owens G, Alterson KD, Lee JS, Groebner JL, Smith JF. Alphavirus replicon approach to promoterless analysis of IRES elements. Virology 2007; 360:376-87. [PMID: 17156813 PMCID: PMC1885372 DOI: 10.1016/j.virol.2006.10.049] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2006] [Revised: 08/30/2006] [Accepted: 10/30/2006] [Indexed: 02/05/2023]
Abstract
Here we describe a system for promoterless analysis of putative internal ribosome entry site (IRES) elements using an alphavirus (family Togaviridae) replicon vector. The system uses the alphavirus subgenomic promoter to produce transcripts that, when modified to contain a spacer region upstream of an IRES element, allow analysis of cap-independent translation of genes of interest (GOI). If the IRES element is removed, translation of the subgenomic transcript can be reduced >95% compared to the same transcript containing a functional IRES element. Alphavirus replicons, used in this manner, offer an alternative to standard dicistronic DNA vectors or in vitro translation systems currently used to analyze putative IRES elements. In addition, protein expression levels varied depending on the spacer element located upstream of each IRES. The ability to modulate the level of expression from alphavirus vectors should extend the utility of these vectors in vaccine development.
Collapse
Affiliation(s)
- K I Kamrud
- AlphaVax, Inc., 2 Triangle Drive, Research Triangle Park, NC 27709-0307, USA.
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Lewis SM, Veyrier A, Hosszu Ungureanu N, Bonnal S, Vagner S, Holcik M. Subcellular relocalization of a trans-acting factor regulates XIAP IRES-dependent translation. Mol Biol Cell 2007; 18:1302-11. [PMID: 17287399 PMCID: PMC1838995 DOI: 10.1091/mbc.e06-06-0515] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Revised: 01/19/2007] [Accepted: 01/31/2007] [Indexed: 01/15/2023] Open
Abstract
Translation of the X-linked inhibitor of apoptosis (XIAP) proceeds by internal ribosome entry site (IRES)-mediated initiation, a process that is physiologically important because XIAP expression is essential for cell survival under conditions of compromised cap-dependent translation, such as cellular stress. The regulation of internal initiation requires the interaction of IRES trans-acting factors (ITAFs) with the IRES element. We used RNA-affinity chromatography to identify XIAP ITAFs and isolated the heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1). We find that hnRNP A1 interacts with XIAP IRES RNA both in vitro and in vivo and that hnRNP A1 negatively regulates XIAP IRES activity. Moreover, XIAP IRES-dependent translation is significantly reduced when hnRNP A1 accumulates in the cytoplasm. Osmotic shock, a cellular stress that causes cytoplasmic accumulation of hnRNP A1, also leads to a decrease in XIAP levels that is abrogated by knockdown of hnRNP A1 expression. These results suggest that the subcellular localization of hnRNP A1 is an important determinant of its ability to negatively regulate XIAP IRES activity, suggesting that the subcellular distribution of ITAFs plays a critical role in regulating IRES-dependent translation. Our findings demonstrate that cytoplasmic hnRNP A1 is a negative regulator of XIAP IRES-dependent translation, indicating a novel function for the cytoplasmic form of this protein.
Collapse
Affiliation(s)
- Stephen M. Lewis
- *Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, K1H 8L1, Canada
| | - Anne Veyrier
- INSERM U563, Toulouse, F-31000, France
- Institut Claudius Régaud, Toulouse, F-31052, France; and
- Université Toulouse III Paul Sabatier, Toulouse, F-31000, France
| | - Nicoleta Hosszu Ungureanu
- *Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, K1H 8L1, Canada
| | - Sophie Bonnal
- INSERM U563, Toulouse, F-31000, France
- Institut Claudius Régaud, Toulouse, F-31052, France; and
- Université Toulouse III Paul Sabatier, Toulouse, F-31000, France
| | - Stéphan Vagner
- INSERM U563, Toulouse, F-31000, France
- Institut Claudius Régaud, Toulouse, F-31052, France; and
- Université Toulouse III Paul Sabatier, Toulouse, F-31000, France
| | - Martin Holcik
- *Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, K1H 8L1, Canada
| |
Collapse
|
99
|
Gerbasi VR, Link AJ. The myotonic dystrophy type 2 protein ZNF9 is part of an ITAF complex that promotes cap-independent translation. Mol Cell Proteomics 2007; 6:1049-58. [PMID: 17327219 DOI: 10.1074/mcp.m600384-mcp200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The 5'-untranslated region of the ornithine decarboxylase (ODC) mRNA contains an internal ribosomal entry site (IRES). Mutational analysis of the ODC IRES has led to the identification of sequences necessary for cap-independent translation of the ODC mRNA. To discover novel IRES trans-acting factors (ITAFs), we performed a proteomics screen for proteins that regulate ODC translation using the wild-type ODC mRNA and a mutant version with an inactive IRES. We identified two RNA-binding proteins that associate with the wild-type ODC IRES but not the mutant IRES. One of these RNA-binding proteins, PCBP2, is an established activator of viral and cellular IRESs. The second protein, ZNF9 (myotonic dystrophy type 2 protein), has not been shown previously to bind IRES-like elements. Using a series of biochemical assays, we validated the interaction of these proteins with ODC mRNA. Interestingly ZNF9 and PCBP2 biochemically associated with each other and appeared to function as part of a larger holo-ITAF ribonucleoprotein complex. Our functional studies showed that PCBP2 and ZNF9 stimulate translation of the ODC IRES. Importantly these results may provide insight into the normal role of ZNF9 and why ZNF9 mutations cause myotonic dystrophy.
Collapse
Affiliation(s)
- Vincent R Gerbasi
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
100
|
Halaby MJ, Yang DQ. p53 translational control: a new facet of p53 regulation and its implication for tumorigenesis and cancer therapeutics. Gene 2007; 395:1-7. [PMID: 17395405 DOI: 10.1016/j.gene.2007.01.029] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Revised: 01/26/2007] [Accepted: 01/26/2007] [Indexed: 01/09/2023]
Abstract
While posttranslational regulation of p53 levels by its interaction with the ubiquitin ligase MDM2 is widely accepted, it has recently become clear that regulation of p53 translation also contributes to p53 induction following DNA damage. However, the mechanisms underlying the translational control of p53 are still poorly understood. In this review, we will focus on the translational regulation of p53 through the 5'- and 3'-untranslated regions of its mRNA. We will also discuss in detail the recent discovery of the p53 internal ribosome entry site (IRES), its role in p53 translation in response to DNA damage, and how it might lead to a better understanding of the process of oncogenesis and provide new avenues for cancer therapeutics.
Collapse
Affiliation(s)
- Marie-Jo Halaby
- Sanford School of Medicine, The University of South Dakota, Division of Basic Biomedical Sciences, 414 East Clark Street, Lee Medicine Building, Vermillion, South Dakota 57069, USA
| | | |
Collapse
|