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










Publication year range
1.
Cell Rep ; 43(6): 114260, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38838225

ABSTRACT

Immunotherapy remains underexploited in acute myeloid leukemia (AML) compared to other hematological malignancies. Currently, gemtuzumab ozogamicin is the only therapeutic antibody approved for this disease. Here, to identify potential targets for immunotherapeutic intervention, we analyze the surface proteome of 100 genetically diverse primary human AML specimens for the identification of cell surface proteins and conduct single-cell transcriptome analyses on a subset of these specimens to assess antigen expression at the sub-population level. Through this comprehensive effort, we successfully identify numerous antigens and markers preferentially expressed by primitive AML cells. Many identified antigens are targeted by therapeutic antibodies currently under clinical evaluation for various cancer types, highlighting the potential therapeutic value of the approach. Importantly, this initiative uncovers AML heterogeneity at the surfaceome level, identifies several antigens and potential primitive cell markers characterizing AML subgroups, and positions immunotherapy as a promising approach to target AML subgroup specificities.


Subject(s)
Immunotherapy , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/pathology , Immunotherapy/methods , Membrane Proteins/metabolism
3.
Blood Adv ; 6(2): 509-514, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34731885

ABSTRACT

Cholesterol homeostasis has been proposed as one mechanism contributing to chemoresistance in AML and hence, inclusion of statins in therapeutic regimens as part of clinical trials in AML has shown encouraging results. Chemical screening of primary human AML specimens by our group led to the identification of lipophilic statins as potent inhibitors of AMLs from a wide range of cytogenetic groups. Genetic screening to identify modulators of the statin response uncovered the role of protein geranylgeranylation and of RAB proteins, coordinating various aspect of vesicular trafficking, in mediating the effects of statins on AML cell viability. We further show that statins can inhibit vesicle-mediated transport in primary human specimens, and that statins sensitive samples show expression signatures reminiscent of enhanced vesicular trafficking. Overall, this study sheds light into the mechanism of action of statins in AML and identifies a novel vulnerability for cytogenetically diverse AML.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Leukemia, Myeloid, Acute , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics
4.
Blood ; 135(21): 1882-1886, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32315381

ABSTRACT

RUNX1 is mutated in ∼10% of adult acute myeloid leukemia (AML). Although most RUNX1 mutations in this disease are believed to be acquired, they can also be germline. Indeed, germline RUNX1 mutations result in the well-described autosomal-dominant familial platelet disorder with predisposition to hematologic malignancies (RUNX1-FPD, FPD/AML, FPDMM); ∼44% of affected individuals progress to AML or myelodysplastic syndromes. Using the Leucegene RUNX1 AML patient group, we sought to investigate the proportion of germline vs acquired RUNX1 mutations in this cohort. Our results showed that 30% of RUNX1 mutations in our AML cohort are germline. Molecular profiling revealed higher frequencies of NRAS mutations and other mutations known to activate various signaling pathways in these patients with RUNX1 germline-mutated AML. Moreover, 2 patients (mother and son) had co-occurrence of RUNX1 and CEBPA germline mutations, with variable AML disease onset at 59 and 27 years, respectively. Together, these data suggest a higher than anticipated frequency of germline RUNX1 mutations in the Leucegene cohort and further highlight the importance of testing for RUNX1 mutations in instances in which allogeneic stem cell transplantation using a related donor is envisioned.


Subject(s)
Biomarkers, Tumor/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Core Binding Factor Alpha 2 Subunit/genetics , GATA2 Transcription Factor/genetics , Germ-Line Mutation , Leukemia, Myeloid, Acute/genetics , Mutation , Adult , Aged , Female , Follow-Up Studies , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Prognosis
5.
Leukemia ; 34(1): 63-74, 2020 01.
Article in English | MEDLINE | ID: mdl-31300747

ABSTRACT

Acute myeloid leukemias (AML) with mutations in the NPM1 gene (NPM1c+) represent a large AML subgroup with varying response to conventional treatment, highlighting the need to develop targeted therapeutic strategies for this disease. We screened a library of clinical drugs on a cohort of primary human AML specimens and identified the BCL2 inhibitor ABT-199 as a selective agent against NPM1c+ AML. Mutational analysis of ABT-199-sensitive and -resistant specimens identified mutations in NPM1, RAD21, and IDH1/IDH2 as predictors of ABT-199 sensitivity. Comparative transcriptome analysis further uncovered BCL2A1 as a potential mediator of ABT-199 resistance in AML. In line with our observation that RAD21 mutation confers sensitivity to ABT-199, we provide functional evidence that reducing RAD21 levels can sensitize AML cells to BCL2 inhibition. Moreover, we demonstrate that ABT-199 is able to produce selective anti-AML activity in vivo toward AML with mutations associated with compound sensitivity in PDX models. Overall, this study delineates the contribution of several genetic events to the response to ABT-199 and provides a rationale for the development of targeted therapies for NPM1c+ AML.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/genetics , Minor Histocompatibility Antigens/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Sulfonamides/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Mutation , Nuclear Proteins/genetics , Nucleophosmin , Tumor Cells, Cultured
6.
Cancer Cell ; 36(1): 84-99.e8, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31287994

ABSTRACT

To identify therapeutic targets in acute myeloid leukemia (AML), we chemically interrogated 200 sequenced primary specimens. Mubritinib, a known ERBB2 inhibitor, elicited strong anti-leukemic effects in vitro and in vivo. In the context of AML, mubritinib functions through ubiquinone-dependent inhibition of electron transport chain (ETC) complex I activity. Resistance to mubritinib characterized normal CD34+ hematopoietic cells and chemotherapy-sensitive AMLs, which displayed transcriptomic hallmarks of hypoxia. Conversely, sensitivity correlated with mitochondrial function-related gene expression levels and characterized a large subset of chemotherapy-resistant AMLs with oxidative phosphorylation (OXPHOS) hyperactivity. Altogether, our work thus identifies an ETC complex I inhibitor and reveals the genetic landscape of OXPHOS dependency in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Oxazoles/pharmacology , Oxidative Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Triazoles/pharmacology , Animals , Biomarkers , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hematopoiesis/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Mice , Models, Biological , Receptor, ErbB-2/antagonists & inhibitors
7.
Cell Rep ; 28(4): 1063-1073.e5, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31340144

ABSTRACT

Transplantation of expanded hematopoietic stem cells (HSCs) and gene therapy based on HSC engineering have emerged as promising approaches for the treatment of hematological diseases. Nevertheless, the immunophenotype of cultured HSCs remains poorly defined. Here, we identify Integrin-α3 (ITGA3) as a marker of cultured human HSCs. Exploiting the pyrimidoindole derivative UM171 to expand cord blood (CB) cells, we show that ITGA3 expression is sufficient to separate the primitive EPCR+CD90+CD133+CD34+CD45RA- HSC population into two functionally distinct fractions presenting mostly short-term (ITGA3-) and both short-term and long-term (ITGA3+) repopulating potential. ITGA3+ cells exhibit robust multilineage differentiation potential, serial reconstitution ability in immunocompromised mice, and an HSC-specific transcriptomic signature. Moreover, ITGA3 expression is functionally required for the long-term engraftment of CB cells. Altogether, our results indicate that ITGA3 is a reliable marker of cultured human long-term repopulating HSCs (LT-HSCs) and represents an important tool to improve the accuracy of prospective HSC identification in culture.


Subject(s)
Biomarkers/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Integrin alpha3/metabolism , Animals , Antigens, CD34/metabolism , Cell Proliferation , Cell Self Renewal , Down-Regulation , Fetal Blood/cytology , Gene Expression Profiling , Humans , Mice , Phenotype , Proto-Oncogene Proteins c-myc/metabolism
8.
Mol Ther Methods Clin Dev ; 10: 156-164, 2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30101153

ABSTRACT

Enhanced gene transfer efficiencies and higher yields of transplantable transduced human hematopoietic stem cells are continuing goals for improving clinical protocols that use stemcell-based gene therapies. Here, we examined the effect of the HSC agonist UM171 on these endpoints in both in vitro and in vivo systems. Using a 22-hr transduction protocol, we found that UM171 significantly enhances both the lentivirus-mediated transduction and yield of CD34+ and CD34+CD45RA- hematopoietic cells from human cord blood to give a 6-fold overall higher recovery of transduced hematopoietic stem cells, including cells with long-term lympho-myeloid repopulating activity in immunodeficient mice. The ability of UM171 to enhance gene transfer to primitive cord blood hematopoietic cells extended to multiple lentiviral pseudotypes, gamma retroviruses, and non-integrating lentiviruses and to adult bone marrow cells. UM171, thus, provides an interesting reagent for improving the ex vivo production of gene-modified cells and for reducing requirements of virus for a broad range of applications.

9.
Clin Cancer Res ; 23(22): 6969-6981, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28855357

ABSTRACT

Purpose:RUNX1-mutated (RUNX1mut) acute myeloid leukemia (AML) is associated with adverse outcome, highlighting the urgent need for a better genetic characterization of this AML subgroup and for the design of efficient therapeutic strategies for this disease. Toward this goal, we further dissected the mutational spectrum and gene expression profile of RUNX1mut AML and correlated these results to drug sensitivity to identify novel compounds targeting this AML subgroup.Experimental Design: RNA-sequencing of 47 RUNX1mut primary AML specimens was performed and sequencing results were compared to those of RUNX1 wild-type samples. Chemical screens were also conducted using RUNX1mut specimens to identify compounds selectively affecting the viability of RUNX1mut AML.Results: We show that samples with no remaining RUNX1 wild-type allele are clinically and genetically distinct and display a more homogeneous gene expression profile. Chemical screening revealed that most RUNX1mut specimens are sensitive to glucocorticoids (GCs) and we confirmed that GCs inhibit AML cell proliferation through their interaction with the glucocorticoid receptor (GR). We observed that specimens harboring RUNX1 mutations expected to result in low residual RUNX1 activity are most sensitive to GCs, and that coassociating mutations as well as GR levels contribute to GC sensitivity. Accordingly, acquired glucocorticoid sensitivity was achieved by negatively regulating RUNX1 expression in human AML cells.Conclusions: Our findings show the profound impact of RUNX1 allele dosage on gene expression profile and glucocorticoid sensitivity in AML, thereby opening opportunities for preclinical testing which may lead to drug repurposing and improved disease characterization. Clin Cancer Res; 23(22); 6969-81. ©2017 AACR.


Subject(s)
Alleles , Core Binding Factor Alpha 2 Subunit/genetics , Drug Resistance, Neoplasm/genetics , Gene Dosage , Glucocorticoids/pharmacology , Leukemia, Myeloid, Acute/genetics , Mutation , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Gene Expression Regulation, Leukemic , Gene Silencing , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged
10.
Blood ; 124(15): 2362-9, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25185265

ABSTRACT

Multipotent long-term repopulating hematopoietic stem cells (LT-HSCs) can self-renew or differentiate into the less primitive short-term repopulating stem cells (ST-HSCs), which themselves produce progenitors that ensure the daily supply of all essential blood components. The Polycomb group (PcG) protein BMI1 is essential for the activity of both HSCs and progenitor cells. Although BMI1 operates by suppressing the Ink4a/Arf locus in progenitors and ST-HSCs, the mechanisms through which this gene regulates the activity of LT-HSCs remain poorly understood. Toward this goal, we isolated BMI1-containing protein complexes and identified UBAP2L as a novel BMI1-interacting protein. We also showed that UBAP2L is preferentially expressed in mouse and human HSC-enriched populations when compared with more mature cell types, and that this gene is essential for the activity of LT-HSCs. In contrast to what is observed for Bmi1 knockdown, we found that UBAP2L depletion does not affect the Ink4a/Arf locus. Given that we demonstrated that BMI1 overexpression is able to rescue the deleterious effects of Ubap2l downregulation on LT-HSC activity and that UBAP2L is part of a PcG subcomplex comprising BMI1, we propose a model in which at least 2 different BMI1-containing PcG complexes regulate HSC activity, which are distinguishable by the presence of UBAP2L.


Subject(s)
Carrier Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Bone Marrow Cells/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation , Gene Deletion , Gene Knockdown Techniques , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Mice, Inbred C57BL , Polycomb-Group Proteins/metabolism , Protein Binding , RNA, Small Interfering/metabolism , Ubiquitin-Protein Ligases/metabolism
11.
PLoS One ; 4(4): e5223, 2009.
Article in English | MEDLINE | ID: mdl-19401772

ABSTRACT

BACKGROUND: Flavaglines are a family of natural products from the genus Aglaia that exhibit anti-cancer activity in vitro and in vivo and inhibit translation initiation. They have been shown to modulate the activity of eIF4A, the DEAD-box RNA helicase subunit of the eukaryotic initiation factor (eIF) 4F complex, a complex that stimulates ribosome recruitment during translation initiation. One flavagline, silvestrol, is capable of modulating chemosensitivity in a mechanism-based mouse model. METHODOLOGY/PRINCIPAL FINDINGS: Among a number of flavagline family members tested herein, we find that silvestrol is the more potent translation inhibitor among these. We find that silvestrol impairs the ribosome recruitment step of translation initiation by affecting the composition of the eukaryotic initiation factor (eIF) 4F complex. We show that silvestrol exhibits significant anticancer activity in human breast and prostate cancer xenograft models, and that this is associated with increased apoptosis, decreased proliferation, and inhibition of angiogenesis. We demonstrate that targeting translation by silvestrol results in preferential inhibition of weakly initiating mRNAs. CONCLUSIONS/SIGNIFICANCE: Our results indicate that silvestrol is a potent anti-cancer compound in vivo that exerts its activity by affecting survival pathways as well as angiogenesis. We propose that silvestrol mediates its effects by preferentially inhibiting translation of malignancy-related mRNAs. Silvestrol appears to be well tolerated in animals.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Triterpenes/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Base Sequence , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Eukaryotic Initiation Factor-4E/metabolism , Female , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/prevention & control , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Transplantation, Heterologous , Triterpenes/chemistry
12.
J Clin Invest ; 118(7): 2651-60, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18551192

ABSTRACT

Disablement of cell death programs in cancer cells contributes to drug resistance and in some cases has been associated with altered translational control. As eukaryotic translation initiation factor 4E (eIF4E) cooperates with c-Myc during lymphomagenesis, induces drug resistance, and is a genetic modifier of the rapamycin response, we have investigated the effect of dysregulation of the ribosome recruitment phase of translation initiation on tumor progression and chemosensitivity. eIF4E is a subunit of eIF4F, a complex that stimulates ribosome recruitment during translation initiation by delivering the DEAD-box RNA helicase eIF4A to the 5' end of mRNAs. eIF4A is thought to prepare a ribosome landing pad on mRNA templates for incoming 40S ribosomes (and associated factors). Using small molecule screening, we found that cyclopenta[b]benzofuran flavaglines, a class of natural products, modulate eIF4A activity and inhibit translation initiation. One member of this class of compounds, silvestrol, was able to enhance chemosensitivity in a mouse lymphoma model in which carcinogenesis is driven by phosphatase and tensin homolog (PTEN) inactivation or elevated eIF4E levels. These results establish that targeting translation initiation can restore drug sensitivity in vivo and provide an approach to modulating chemosensitivity.


Subject(s)
Benzofurans/pharmacology , Drug Resistance, Neoplasm/drug effects , Lymphoma/drug therapy , Peptide Chain Initiation, Translational/drug effects , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Disease Models, Animal , Doxorubicin/pharmacology , Drug Synergism , Eukaryotic Initiation Factor-4A/analysis , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Female , HeLa Cells , Humans , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Polyribosomes/drug effects , Polyribosomes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/pharmacology , Thapsigargin/pharmacology
13.
PLoS One ; 3(2): e1583, 2008 Feb 13.
Article in English | MEDLINE | ID: mdl-18270573

ABSTRACT

RNA helicases represent a large family of proteins implicated in many biological processes including ribosome biogenesis, splicing, translation and mRNA degradation. However, these proteins have little substrate specificity, making inhibition of selected helicases a challenging problem. The prototypical DEAD box RNA helicase, eIF4A, works in conjunction with other translation factors to prepare mRNA templates for ribosome recruitment during translation initiation. Herein, we provide insight into the selectivity of a small molecule inhibitor of eIF4A, hippuristanol. This coral-derived natural product binds to amino acids adjacent to, and overlapping with, two conserved motifs present in the carboxy-terminal domain of eIF4A. Mutagenesis of amino acids within this region allowed us to alter the hippuristanol-sensitivity of eIF4A and undertake structure/function studies. Our results provide an understanding into how selective targeting of RNA helicases for pharmacological intervention can be achieved.


Subject(s)
DEAD-box RNA Helicases/metabolism , Drug Delivery Systems/methods , Sterols/pharmacology , Animals , Binding Sites , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4A , Mice , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Sterols/chemistry
14.
Chem Biol ; 13(12): 1287-95, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17185224

ABSTRACT

Eukaryotic initiation factor 4A (eIF4A) is a member of the DEAD-box family of putative RNA helicases whose members are involved in many aspects of RNA metabolism. eIF4A is thought to facilitate binding of 43S preinitiation complexes to mRNAs by unwinding secondary structures present in the 5' untranslated region. Pateamine A, a small-molecule inhibitor of translation initiation, acts in an unusual manner by stimulating eIF4A activity. Herein, we report the elucidation of pateamine's mode of action. We demonstrate that Pateamine A is a chemical inducer of dimerization that forces an engagement between eIF4A and RNA and prevents eIF4A from participating in the ribosome-recruitment step of translation initiation.


Subject(s)
Epoxy Compounds/pharmacology , Eukaryotic Initiation Factor-4A/drug effects , Eukaryotic Initiation Factor-4A/metabolism , Macrolides/pharmacology , Protein Biosynthesis/physiology , RNA/metabolism , Thiazoles/pharmacology , Dimerization , Eukaryotic Initiation Factor-4G/drug effects , Eukaryotic Initiation Factor-4G/metabolism , Humans , Jurkat Cells , Multiprotein Complexes/drug effects , Multiprotein Complexes/metabolism , Protein Binding/physiology , Protein Biosynthesis/drug effects
15.
Mol Biol Cell ; 17(10): 4212-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16870703

ABSTRACT

Cytoplasmic aggregates known as stress granules (SGs) arise as a consequence of cellular stress and contain stalled translation preinitiation complexes. These foci are thought to serve as sites of mRNA storage or triage during the cell stress response. SG formation has been shown to require induction of eukaryotic initiation factor (eIF)2alpha phosphorylation. Herein, we investigate the potential role of other initiation factors in this process and demonstrate that interfering with eIF4A activity, an RNA helicase required for the ribosome recruitment phase of translation initiation, induces SG formation and that this event is not dependent on eIF2alpha phosphorylation. We also show that inhibition of eIF4A activity does not impair the ability of eIF2alpha to be phosphorylated under stress conditions. Furthermore, we observed SG assembly upon inhibition of cap-dependent translation after poliovirus infection. We propose that SG modeling can occur via both eIF2alpha phosphorylation-dependent and -independent pathways that target translation initiation.


Subject(s)
Cytoplasmic Granules/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4A/physiology , Peptide Initiation Factors/metabolism , Ribosomes/metabolism , Antigens, Surface/metabolism , Carrier Proteins/metabolism , DNA Helicases , ELAV Proteins , ELAV-Like Protein 1 , Eukaryotic Initiation Factor-2/physiology , Eukaryotic Initiation Factor-4A/genetics , HeLa Cells , Humans , Models, Genetic , Phosphorylation , Poliovirus/genetics , Poly-ADP-Ribose Binding Proteins , Protein Biosynthesis , RNA Helicases , RNA Recognition Motif Proteins , RNA, Small Interfering , RNA-Binding Proteins/metabolism
16.
J Biol Chem ; 281(35): 25315-25, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16835235

ABSTRACT

Two classes of viruses, namely members of the Potyviridae and Caliciviridae, use a novel mechanism for the initiation of protein synthesis that involves the interaction of translation initiation factors with a viral protein covalently linked to the viral RNA, known as VPg. The calicivirus VPg proteins can interact directly with the initiation factors eIF4E and eIF3. Translation initiation on feline calicivirus (FCV) RNA requires eIF4E because it is inhibited by recombinant 4E-BP1. However, to date, there have been no functional studies carried out with respect to norovirus translation initiation, because of a lack of a suitable source of VPg-linked viral RNA. We have now used the recently identified murine norovirus (MNV) as a model system for norovirus translation and have extended our previous studies with FCV RNA to examine the role of the other eIF4F components in translation initiation. We now demonstrate that, as with FCV, MNV VPg interacts directly with eIF4E, although, unlike FCV RNA, translation of MNV RNA is not sensitive to 4E-BP1, eIF4E depletion, or foot-and-mouth disease virus Lb protease-mediated cleavage of eIF4G. We also demonstrate that both FCV and MNV RNA translation require the RNA helicase component of the eIF4F complex, namely eIF4A, because translation was sensitive (albeit to different degrees) to a dominant negative form and to a small molecule inhibitor of eIF4A (hippuristanol). These results suggest that calicivirus RNAs differ with respect to their requirements for the components of the eIF4F translation initiation complex.


Subject(s)
Caliciviridae/physiology , Eukaryotic Initiation Factor-4F/chemistry , Gene Expression Regulation, Viral , Animals , Genes, Dominant , Mice , Mutation , Norovirus/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Rabbits , Recombinant Proteins/chemistry , Sterols/chemistry
17.
J Gen Virol ; 87(Pt 4): 927-936, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16528042

ABSTRACT

The internal ribosome entry site (IRES) elements from porcine enterovirus 8 and simian virus 2, two members of a proposed new genus within the family Picornaviridae, were characterized. These IRES elements, in common with the porcine teschovirus 1 IRES, were found to be related functionally and structurally to the IRES element from Hepatitis C virus, a member of the family Flaviviridae. Partial secondary structure predictions were derived and functional assays demonstrated that these IRES elements continued to be active when eIF4G was cleaved and when the activity of eIF4A was blocked.


Subject(s)
Enteroviruses, Porcine/genetics , Hepacivirus/genetics , Picornaviridae/genetics , Picornaviridae/pathogenicity , Ribosomes/metabolism , 5' Untranslated Regions/chemistry , 5' Untranslated Regions/genetics , Animals , Base Sequence , Codon, Initiator , DNA, Complementary/chemistry , DNA, Complementary/genetics , Enteroviruses, Porcine/pathogenicity , Hepacivirus/pathogenicity , Humans , Molecular Sequence Data , Picornaviridae/classification , RNA, Viral/chemistry , RNA, Viral/genetics , Swine/virology
18.
Nat Chem Biol ; 2(4): 213-20, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16532013

ABSTRACT

RNA helicases are molecular motors that are involved in virtually all aspects of RNA metabolism. Eukaryotic initiation factor (eIF) 4A is the prototypical member of the DEAD-box family of RNA helicases. It is thought to use energy from ATP hydrolysis to unwind mRNA structure and, in conjunction with other translation factors, it prepares mRNA templates for ribosome recruitment during translation initiation. In screening marine extracts for new eukaryotic translation initiation inhibitors, we identified the natural product hippuristanol. We show here that this compound is a selective and potent inhibitor of eIF4A RNA-binding activity that can be used to distinguish between eIF4A-dependent and -independent modes of translation initiation in vitro and in vivo. We also show that poliovirus replication is delayed when infected cells are exposed to hippuristanol. Our study demonstrates the feasibility of selectively targeting members of the DEAD-box helicase family with small-molecule inhibitors.


Subject(s)
Eukaryotic Initiation Factor-4A/chemistry , RNA Helicases/chemistry , Ribosomes/chemistry , Adenosine Triphosphate/chemistry , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Humans , Hydrolysis , Magnetic Resonance Spectroscopy , Models, Genetic , Plasmids/metabolism , Poliovirus/genetics , Poliovirus/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Biosynthesis , RNA/chemistry , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Structure-Activity Relationship , Transfection
19.
Mol Cell ; 21(4): 533-42, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16483934

ABSTRACT

MicroRNAs (miRNAs) are predicted to regulate 30% of mammalian protein-encoding genes by interactions with their 3' untranslated regions (UTRs). We use partially complementary siRNAs to investigate the mechanism by which miRNAs mediate translational repression in human cells. Repressed mRNAs are associated with polyribosomes that are engaged in translation elongation, as shown by puromycin sensitivity. The inhibition appears to be postinitiation because translation driven by the cap-independent processes of HCV IRES and CrPV IRES is repressed by short RNAs. Further, metabolic labeling suggests that silencing occurs before completion of the nascent polypeptide chain. In addition, silencing by short RNAs causes a decrease in translational readthrough at a stop codon, and ribosomes on repressed mRNAs dissociate more rapidly after a block of initiation of translation than those on control mRNAs. These results suggest that repression by short RNAs, and thus probably miRNAs, is primarily due to ribosome drop off during elongation of translation.


Subject(s)
Gene Silencing , MicroRNAs/metabolism , Protein Biosynthesis , Animals , Cell Line , Codon, Terminator , Gene Expression Regulation , Humans , Peptide Chain Termination, Translational , Peptides/metabolism , Peptidyl Transferases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribosomes/metabolism
20.
Proc Natl Acad Sci U S A ; 102(30): 10460-5, 2005 Jul 26.
Article in English | MEDLINE | ID: mdl-16030146

ABSTRACT

RNA helicases are the largest group of enzymes in eukaryotic RNA metabolism. The DEXD/H-box putative RNA helicases form the helicase superfamily II, whose members are defined by seven highly conserved amino acid motifs, making specific targeting of selected members a challenging pharmacological problem. The translation initiation factor eIF4A is the prototypical DEAD-box RNA helicase that works in conjunction with eIF4B and eIF4H and as a subunit of eIF4F to prepare the mRNA template for ribosome binding, possibly by unwinding the secondary structure proximal to the 5' m7GpppN cap structure. We report the identification and characterization of a small molecule inhibitor of eukaryotic translation initiation that acts in an unusual manner by stimulating eIF4A-associated activities. Our results suggest that proper control of eIF4A helicase activity is necessary for efficient ribosome binding and demonstrate the feasibility of selectively targeting DEAD-box RNA helicases with small molecules.


Subject(s)
Epoxy Compounds/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Ribosomes/metabolism , Thiazoles/metabolism , Animals , Chromatography, Agarose , Dose-Response Relationship, Drug , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Escherichia coli , Macrolides , Mice , Thiazoles/chemistry , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...