Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 619
Filter
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35064089

ABSTRACT

Kasugamycin (KSG) is an aminoglycoside antibiotic widely used in agriculture and exhibits considerable medical potential. Previous studies suggested that KSG interferes with translation by blocking binding of canonical messenger RNA (mRNA) and initiator transfer tRNA (tRNA) to the small ribosomal subunit, thereby preventing initiation of protein synthesis. Here, by using genome-wide approaches, we show that KSG can interfere with translation even after the formation of the 70S initiation complex on mRNA, as the extent of KSG-mediated translation inhibition correlates with increased occupancy of start codons by 70S ribosomes. Even at saturating concentrations, KSG does not completely abolish translation, allowing for continuing expression of some Escherichia coli proteins. Differential action of KSG significantly depends on the nature of the mRNA residue immediately preceding the start codon, with guanine in this position being the most conducive to inhibition by the drug. In addition, the activity of KSG is attenuated by translational coupling as genes whose start codons overlap with the coding regions or the stop codons of the upstream cistrons tend to be less susceptible to drug-mediated inhibition. Altogether, our findings reveal KSG as an example of a small ribosomal subunit-targeting antibiotic with a well-pronounced context specificity of action.


Subject(s)
Aminoglycosides/pharmacology , Binding Sites , Peptide Chain Initiation, Translational/drug effects , RNA, Messenger/genetics , Ribosomes/metabolism , Aminoglycosides/chemistry , Codon, Initiator , Molecular Structure , Open Reading Frames , Protein Binding , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Ribosomes/chemistry , Structure-Activity Relationship
2.
RNA ; 27(10): 1241-1256, 2021 10.
Article in English | MEDLINE | ID: mdl-34244458

ABSTRACT

Stress granules (SGs) are membraneless organelles composed of mRNAs and RNA binding proteins which undergo assembly in response to stress-induced inactivation of translation initiation. In general, SG recruitment is limited to a subpopulation of a given mRNA species and RNA-seq analyses of purified SGs revealed that signal sequence-encoding (i.e., endoplasmic reticulum [ER]-targeted) transcripts are significantly underrepresented, consistent with prior reports that ER localization can protect mRNAs from SG recruitment. Using translational profiling, cell fractionation, and single molecule mRNA imaging, we examined SG biogenesis following activation of the unfolded protein response (UPR) by 1,4-dithiothreitol (DTT) and report that gene-specific subsets of cytosolic and ER-targeted mRNAs can be recruited into SGs. Furthermore, we demonstrate that SGs form in close proximity to or directly associated with the ER membrane. ER-associated SG assembly was also observed during arsenite stress, suggesting broad roles for the ER in SG biogenesis. Recruitment of a given mRNA into SGs required stress-induced translational repression, though translational inhibition was not solely predictive of an mRNA's propensity for SG recruitment. SG formation was prevented by the transcriptional inhibitors actinomycin D or triptolide, suggesting a functional link between gene transcriptional state and SG biogenesis. Collectively these data demonstrate that ER-targeted and cytosolic mRNAs can be recruited into ER-associated SGs and this recruitment is sensitive to transcriptional inhibition. We propose that newly transcribed mRNAs exported under conditions of suppressed translation initiation are primary SG substrates, with the ER serving as the central subcellular site of SG formation.


Subject(s)
Cytoplasmic Granules/genetics , Endoplasmic Reticulum/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Unfolded Protein Response , Biomarkers/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Cytosol/drug effects , Cytosol/metabolism , Dactinomycin/pharmacology , Diterpenes/pharmacology , Dithiothreitol/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Epoxy Compounds/pharmacology , Gene Expression , HeLa Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Organelle Biogenesis , Peptide Chain Initiation, Translational/drug effects , Phenanthrenes/pharmacology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Single Molecule Imaging , Stress, Physiological/drug effects , Transcription, Genetic/drug effects , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism
3.
Molecules ; 26(11)2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34200016

ABSTRACT

The increase in antibacterial resistance is a serious challenge for both the health and defence sectors and there is a need for both novel antibacterial targets and antibacterial strategies. RNA degradation and ribonucleases, such as the essential endoribonuclease RNase E, encoded by the rne gene, are emerging as potential antibacterial targets while antisense oligonucleotides may provide alternative antibacterial strategies. As rne mRNA has not been previously targeted using an antisense approach, we decided to explore using antisense oligonucleotides to target the translation initiation region of the Escherichia coli rne mRNA. Antisense oligonucleotides were rationally designed and were synthesised as locked nucleic acid (LNA) gapmers to enable inhibition of rne mRNA translation through two mechanisms. Either LNA gapmer binding could sterically block translation and/or LNA gapmer binding could facilitate RNase H-mediated cleavage of the rne mRNA. This may prove to be an advantage over the majority of previous antibacterial antisense oligonucleotide approaches which used oligonucleotide chemistries that restrict the mode-of-action of the antisense oligonucleotide to steric blocking of translation. Using an electrophoretic mobility shift assay, we demonstrate that the LNA gapmers bind to the translation initiation region of E. coli rne mRNA. We then use a cell-free transcription translation reporter assay to show that this binding is capable of inhibiting translation. Finally, in an in vitro RNase H cleavage assay, the LNA gapmers facilitate RNase H-mediated mRNA cleavage. Although the challenges of antisense oligonucleotide delivery remain to be addressed, overall, this work lays the foundations for the development of a novel antibacterial strategy targeting rne mRNA with antisense oligonucleotides.


Subject(s)
Anti-Bacterial Agents/pharmacology , Endoribonucleases/genetics , Escherichia coli/enzymology , Oligonucleotides/pharmacology , Cell-Free System , Endoribonucleases/antagonists & inhibitors , Escherichia coli/drug effects , Escherichia coli Proteins/genetics , Oligonucleotides/chemical synthesis , Peptide Chain Initiation, Translational/drug effects , RNA, Messenger/antagonists & inhibitors
4.
Am J Physiol Endocrinol Metab ; 320(3): E551-E565, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33427053

ABSTRACT

Extrauterine growth restriction in premature infants is largely attributed to reduced lean mass accretion and is associated with long-term morbidities. Previously, we demonstrated that prematurity blunts the feeding-induced stimulation of translation initiation signaling and protein synthesis in skeletal muscle of neonatal pigs. The objective of the current study was to determine whether the blunted feeding response is mediated by reduced responsiveness to insulin, amino acids, or both. Pigs delivered by cesarean section preterm (PT; 103 days, n = 25) or at term (T; 112 days, n = 26) were subject to euinsulinemic-euaminoacidemic-euglycemic (FAST), hyperinsulinemic-euaminoacidemic-euglycemic (INS), or euinsulinemic-hyperaminoacidemic-euglycemic (AA) clamps four days after delivery. Indices of mechanistic target of rapamycin complex 1 (mTORC1) signaling and fractional protein synthesis rates were measured after 2 h. Although longissimus dorsi (LD) muscle protein synthesis increased in response to both INS and AA, the increase was 28% lower in PT than in T. Upstream of mTORC1, Akt phosphorylation, an index of insulin signaling, was increased with INS but was 40% less in PT than in T. The abundances of mTOR·RagA and mTOR·RagC, indices of amino acid signaling, increased with AA but were 25% less in PT than in T. Downstream of mTORC1, eIF4E·eIF4G abundance was increased by both INS and AA but attenuated by prematurity. These results suggest that preterm birth blunts both insulin- and amino acid-induced activation of mTORC1 and protein synthesis in skeletal muscle, thereby limiting the anabolic response to feeding. This anabolic resistance likely contributes to the high prevalence of extrauterine growth restriction in prematurity.NEW & NOTEWORTHY Extrauterine growth faltering is a major complication of premature birth, but the underlying cause is poorly understood. Our results demonstrate that preterm birth blunts both the insulin-and amino acid-induced activation of mTORC1-dependent translation initiation and protein synthesis in skeletal muscle, thereby limiting the anabolic response to feeding. This anabolic resistance likely contributes to the reduced accretion of lean mass and extrauterine growth restriction of premature infants.


Subject(s)
Amino Acids/pharmacology , Insulin/pharmacology , Muscle, Skeletal/drug effects , Peptide Chain Initiation, Translational/drug effects , Premature Birth/metabolism , Protein Biosynthesis/drug effects , Amino Acids/metabolism , Animals , Animals, Newborn , Female , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Muscle, Skeletal/metabolism , Pregnancy , Signal Transduction/drug effects , Swine
5.
J Invest Dermatol ; 141(4): 742-753.e10, 2021 04.
Article in English | MEDLINE | ID: mdl-32971126

ABSTRACT

Skin squamous cell carcinomas (SCCs) are a major cause of death in patients who have undergone or will undergo organ transplantation. Moreover, these neoplasms cause significant disease and economic burden and diminish patients' life quality. However, no effective treatment or intervention strategies are available. In this study, we investigated the pathologic role of 5'-cap translation, which is regulated by the formation of a ternary initiation factor complex involving eIF4E, eIF4G, and eIF4A1. We detected increased expression of phosphorylated eIF4E, eIF4G, and eIF4A1 in human and murine skin SCCs. The increase in these ternary initiation factor complex proteins was associated with enhanced eIF4E translation targets cyclin D1 and c-Myc. Conversely, small interfering RNA-mediated depletion of eIF4E in human SCC cells (A431 and SCC-13) reduced eIF4G and proteins that regulate the cell cycle and proliferation. Notably, inhibition of Raf/MAPK/extracellular signal-regulated kinase signaling decreased eIF4E and phosphorylated eIF4E accumulation and significantly diminished cell-cycle gene expression and tumor volume of A431-derived xenograft tumors. Furthermore, disrupting the eIF4E with an allosteric inhibitor of eIF4E and eIF4G binding, 4EGI-1, decreased the eIF4E/eIF4G expression and reduced the proliferation. Finally, combined inhibition of the Raf/MAPK/extracellular signal-regulated kinase axis and eIF4E impaired 5'-cap‒dependent translation and abrogated tumor cell proliferation. These data demonstrate that 5'-cap‒dependent translation is a potential therapeutic target for abrogating lethal skin SCCs in patients who have undergone or will undergo organ transplantation.


Subject(s)
Carcinoma, Squamous Cell/genetics , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , RNA, Small Interfering/pharmacology , Skin Neoplasms/genetics , Allosteric Regulation/drug effects , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclin D1/genetics , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Peptide Chain Initiation, Translational/drug effects , Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , RNA Caps/metabolism , RNA, Small Interfering/therapeutic use , Skin/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Nucleic Acids Res ; 48(10): 5201-5216, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32382758

ABSTRACT

High-throughput methods, such as ribosome profiling, have revealed the complexity of translation regulation in Bacteria and Eukarya with large-scale effects on cellular functions. In contrast, the translational landscape in Archaea remains mostly unexplored. Here, we developed ribosome profiling in a model archaeon, Haloferax volcanii, elucidating, for the first time, the translational landscape of a representative of the third domain of life. We determined the ribosome footprint of H. volcanii to be comparable in size to that of the Eukarya. We linked footprint lengths to initiating and elongating states of the ribosome on leadered transcripts, operons, and on leaderless transcripts, the latter representing 70% of H. volcanii transcriptome. We manipulated ribosome activity with translation inhibitors to reveal ribosome pausing at specific codons. Lastly, we found that the drug harringtonine arrested ribosomes at initiation sites in this archaeon. This drug treatment allowed us to confirm known translation initiation sites and also reveal putative novel initiation sites in intergenic regions and within genes. Ribosome profiling revealed an uncharacterized complexity of translation in this archaeon with bacteria-like, eukarya-like, and potentially novel translation mechanisms. These mechanisms are likely to be functionally essential and to contribute to an expanded proteome with regulatory roles in gene expression.


Subject(s)
Codon/metabolism , Haloferax volcanii/genetics , Haloferax volcanii/metabolism , Protein Biosynthesis , Ribosomes/metabolism , 5' Untranslated Regions/genetics , Codon/genetics , Haloferax volcanii/drug effects , Harringtonines/pharmacology , Peptide Chain Elongation, Translational/drug effects , Peptide Chain Elongation, Translational/genetics , Peptide Chain Initiation, Translational/drug effects , Peptide Chain Initiation, Translational/genetics , Protein Biosynthesis/drug effects , Protein Footprinting , Reading Frames/genetics , Ribosomes/drug effects , Transcriptome/drug effects
7.
Bioorg Chem ; 96: 103583, 2020 03.
Article in English | MEDLINE | ID: mdl-31972465

ABSTRACT

Herein we describe a synthesis of new isoxazole-containing 5' mRNA cap analogues via a cycloaddition reaction. The obtained analogues show a capability to inhibit cap-dependent translation in vitro and are characterized by a new binding mode in which an isoxazolic ring, instead of guanine, is involved in the stacking effect. Our study provides valuable information toward designing new compounds that can be potentially used as anticancer therapeutics.


Subject(s)
Isoxazoles/chemistry , Isoxazoles/pharmacology , Peptide Chain Initiation, Translational/drug effects , RNA Cap Analogs/chemistry , RNA Cap Analogs/pharmacology , Animals , Drug Design , Eukaryotic Initiation Factor-4E/metabolism , Isoxazoles/chemical synthesis , Mice , Molecular Docking Simulation , RNA Cap Analogs/chemical synthesis , Rabbits
8.
Cells ; 9(1)2020 01 10.
Article in English | MEDLINE | ID: mdl-31936702

ABSTRACT

Eukaryotic initiation factor 6 (eIF6) is necessary for the nucleolar biogenesis of 60S ribosomes. However, most of eIF6 resides in the cytoplasm, where it acts as an initiation factor. eIF6 is necessary for maximal protein synthesis downstream of growth factor stimulation. eIF6 is an antiassociation factor that binds 60S subunits, in turn preventing premature 40S joining and thus the formation of inactive 80S subunits. It is widely thought that eIF6 antiassociation activity is critical for its function. Here, we exploited and improved our assay for eIF6 binding to ribosomes (iRIA) in order to screen for modulators of eIF6 binding to the 60S. Three compounds, eIFsixty-1 (clofazimine), eIFsixty-4, and eIFsixty-6 were identified and characterized. All three inhibit the binding of eIF6 to the 60S in the micromolar range. eIFsixty-4 robustly inhibits cell growth, whereas eIFsixty-1 and eIFsixty-6 might have dose- and cell-specific effects. Puromycin labeling shows that eIF6ixty-4 is a strong global translational inhibitor, whereas the other two are mild modulators. Polysome profiling and RT-qPCR show that all three inhibitors reduce the specific translation of well-known eIF6 targets. In contrast, none of them affect the nucleolar localization of eIF6. These data provide proof of principle that the generation of eIF6 translational modulators is feasible.


Subject(s)
Peptide Initiation Factors/metabolism , Protein Biosynthesis , Ribosome Subunits, Large, Eukaryotic/metabolism , Cell Line , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Cell Survival , Enzyme-Linked Immunosorbent Assay , Humans , Peptide Chain Initiation, Translational/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Binding/drug effects , Puromycin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results
9.
PLoS Biol ; 18(1): e3000593, 2020 01.
Article in English | MEDLINE | ID: mdl-31995552

ABSTRACT

During host colonization, bacteria use the alarmones (p)ppGpp to reshape their proteome by acting pleiotropically on DNA, RNA, and protein synthesis. Here, we elucidate how the initiating ribosome senses the cellular pool of guanosine nucleotides and regulates the progression towards protein synthesis. Our results show that the affinity of guanosine triphosphate (GTP) and the inhibitory concentration of ppGpp for the 30S-bound initiation factor IF2 vary depending on the programmed mRNA. The TufA mRNA enhanced GTP affinity for 30S complexes, resulting in improved ppGpp tolerance and allowing efficient protein synthesis. Conversely, the InfA mRNA allowed ppGpp to compete with GTP for IF2, thus stalling 30S complexes. Structural modeling and biochemical analysis of the TufA mRNA unveiled a structured enhancer of translation initiation (SETI) composed of two consecutive hairpins proximal to the translation initiation region (TIR) that largely account for ppGpp tolerance under physiological concentrations of guanosine nucleotides. Furthermore, our results show that the mechanism enhancing ppGpp tolerance is not restricted to the TufA mRNA, as similar ppGpp tolerance was found for the SETI-containing Rnr mRNA. Finally, we show that IF2 can use pppGpp to promote the formation of 30S initiation complexes (ICs), albeit requiring higher factor concentration and resulting in slower transitions to translation elongation. Altogether, our data unveil a novel regulatory mechanism at the onset of protein synthesis that tolerates physiological concentrations of ppGpp and that bacteria can exploit to modulate their proteome as a function of the nutritional shift happening during stringent response and infection.


Subject(s)
Guanosine Tetraphosphate/pharmacology , Peptide Chain Initiation, Translational , RNA, Messenger/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Binding, Competitive , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Guanosine Tetraphosphate/metabolism , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Host-Pathogen Interactions/physiology , Kinetics , Nucleic Acid Conformation , Peptide Chain Initiation, Translational/drug effects , Peptide Chain Initiation, Translational/physiology , Peptide Elongation Factor Tu/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/chemistry , RNA, Messenger/drug effects , RNA, Messenger/genetics
10.
Leukemia ; 34(1): 138-150, 2020 01.
Article in English | MEDLINE | ID: mdl-31171817

ABSTRACT

MYC-driven lymphomas, especially those with concurrent MYC and BCL2 dysregulation, are currently a challenge in clinical practice due to rapid disease progression, resistance to standard chemotherapy, and high risk of refractory disease. MYC plays a central role by coordinating hyperactive protein synthesis with upregulated transcription in order to support rapid proliferation of tumor cells. Translation initiation inhibitor rocaglates have been identified as the most potent drugs in MYC-driven lymphomas as they efficiently inhibit MYC expression and tumor cell viability. We found that this class of compounds can overcome eIF4A abundance by stabilizing target mRNA-eIF4A interaction that directly prevents translation. Proteome-wide quantification demonstrated selective repression of multiple critical oncoproteins in addition to MYC in B-cell lymphoma including NEK2, MCL1, AURKA, PLK1, and several transcription factors that are generally considered undruggable. Finally, (-)-SDS-1-021, the most promising synthetic rocaglate, was confirmed to be highly potent as a single agent, and displayed significant synergy with the BCL2 inhibitor ABT199 in inhibiting tumor growth and survival in primary lymphoma cells in vitro and in patient-derived xenograft mouse models. Overall, our findings support the strategy of using rocaglates to target oncoprotein synthesis in MYC-driven lymphomas.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Lymphoma, B-Cell , Peptide Chain Initiation, Translational/drug effects , Proto-Oncogene Proteins c-myc/drug effects , Aglaia , Animals , Female , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Mice , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Xenograft Model Antitumor Assays
11.
Biomolecules ; 9(11)2019 10 29.
Article in English | MEDLINE | ID: mdl-31671902

ABSTRACT

Translation of mRNA is an important process that controls cell behavior and gene regulation because proteins are the functional molecules that determine cell types and function. Cancer develops as a result of genetic mutations, which lead to the production of abnormal proteins and the dysregulation of translation, which in turn, leads to aberrant protein synthesis. In addition, the machinery that is involved in protein synthesis plays critical roles in stem cell fate determination. In the current review, recent advances in the understanding of translational control, especially translational initiation in cancer development and stem cell fate control, are described. Therapeutic targets of mRNA translation such as eIF4E, 4EBP, and eIF2, for cancer treatment or stem cell fate regulation are reviewed. Upstream signaling pathways that regulate and affect translation initiation were introduced. It is important to regulate the expression of protein for normal cell behavior and development. mRNA translation initiation is a key step to regulate protein synthesis, therefore, identifying and targeting molecules that are critical for protein synthesis is necessary and beneficial to develop cancer therapeutics and stem cells fate regulation.


Subject(s)
Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Peptide Chain Initiation, Translational/drug effects , Humans , Neoplasms/genetics , Neoplasms/pathology , RNA, Messenger/genetics
12.
Sci Rep ; 9(1): 1265, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718665

ABSTRACT

Translation is a highly regulated process that is perturbed in human cancers, often through activation of the PI3K/mTOR pathway which impacts directly on the ribosome recruitment phase of translation initiation. While significant research has focused on "drugging" components of the PI3K/mTOR network, efforts have also been directed towards inhibiting eukaryotic initiation factor (eIF) 4F-dependent translation. Small molecule inhibitors of this complex have been identified, characterized, and used to validate the rationale of targeting this step to curtail tumor cell growth and modulate chemotherapy response. One such class of compounds are the rocaglates, secondary metabolites from the plant genus Aglaia, which target the RNA helicase subunit of eIF4F, eIF4A. Here we explore the ability of synthetic derivatives of aglaiastatins and an aglaroxin derivative to target the translation process in vitro and in vivo and find the synthetic derivative oxo-aglaiastatin to possess such activity. Oxo-aglaiastatin inhibited translation in vitro and in vivo and synergized with doxorubicin, ABT-199 (a Bcl-2 antagonist), and dexamethasone when tested on hematological cancer cells. The biological activity of oxo-aglaiastatin was shown to be a consequence of inhibiting eIF4A1 activity.


Subject(s)
Aglaia , Antineoplastic Agents, Phytogenic/pharmacology , Neoplasms/drug therapy , Peptide Chain Initiation, Translational/drug effects , Aglaia/chemistry , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Synergism , Eukaryotic Initiation Factor-4A/metabolism , Female , Humans , Lymphoma/drug therapy , Mice, Inbred C57BL , Neoplasms/metabolism , Sulfonamides/pharmacology
13.
Transl Res ; 207: 83-95, 2019 05.
Article in English | MEDLINE | ID: mdl-30738861

ABSTRACT

Multiple myeloma (MM) malignant plasma cells accumulate in the bone marrow (BM) where their interaction with the microenvironment promotes disease progression and drug resistance. Previously, we have shown that MM cells cocultured with BM-mesenchymal stem cells (MSCs) comodulated cells' phenotype in a MAPKs/translation initiation (TI)-dependent manner. Dissection of the coculture model showed that BM-MSCs secretomes and microvesicles (MVs) participate in this crosstalk. Here, we addressed the role of the BM-MSCs extracellular matrix (ECM). MM cell lines cultured on decellularized ECM of normal donors' (ND) or MM patients' BM-MSCs were assayed for phenotype (viability, cell count, death, proliferation, migration, and invasion), microRNAs (MIR125a-3p, MIR199a-3p) and targets, MAPKs, TI epithelial-to-mesenchymal transition (EMT), CXCR4, and autophagy. Drug (doxorubicin, velcade) response of MM cells cultured on ND/MM-MSCs' ECM with/without adhered MVs was also evaluated. ECM evoked opposite responses according to its origin: MM cells cultured on ND-MSCs' ECM demonstrated a rapid and continued decrease in MAPK/TI activation (↓10%-25%, P < 0.05) (15-24 hours) followed by diminished viability, cell count, proliferation, migration, and invasion (16-72 hours) (↓10%-50%, P < 0.05). In contrast, MM cells cultured on MM-MSCs' ECM displayed activated MAPK/TI, proliferation, EMT, and CXCR4 (↑15%-250%, P < 0.05). Corresponding changes in microRNAs relevant to the MM cells' altered phenotype were also determined. The hierarchy and interdependence of MAPKs/TI/autophagy/phenotype cascade were demonstrated. Finally, we showed that the ECM cooperates with MVs to modulate MM cells drug response. These data demonstrate the contribution of BM-MSCs' ECM to MM niche design and underscore the clinical potential of identifying targetable signals.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Marrow Cells/metabolism , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Bone Marrow Cells/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell-Derived Microparticles/drug effects , Cell-Derived Microparticles/metabolism , Drug Resistance, Neoplasm/drug effects , Extracellular Matrix/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mesenchymal Stem Cells/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Multiple Myeloma/genetics , Neoplasm Invasiveness , Peptide Chain Initiation, Translational/drug effects , Phenotype , Reproducibility of Results
14.
EMBO Rep ; 19(12)2018 12.
Article in English | MEDLINE | ID: mdl-30361391

ABSTRACT

Canonical Wnt/ß-catenin signaling is an essential regulator of various cellular functions throughout development and adulthood. Aberrant Wnt/ß-catenin signaling also contributes to various pathologies including cancer, necessitating an understanding of cell context-dependent mechanisms regulating this pathway. Since protein-protein interactions underpin ß-catenin function and localization, we sought to identify novel ß-catenin interacting partners by affinity purification coupled with tandem mass spectrometry in vascular smooth muscle cells (VSMCs), where ß-catenin is involved in both physiological and pathological control of cell proliferation. Here, we report novel components of the VSMC ß-catenin interactome. Bioinformatic analysis of the protein networks implies potentially novel functions for ß-catenin, particularly in mRNA translation, and we confirm a direct interaction between ß-catenin and the fragile X mental retardation protein (FMRP). Biochemical studies reveal a basal recruitment of ß-catenin to the messenger ribonucleoprotein and translational pre-initiation complex, fulfilling a translational repressor function. Wnt stimulation antagonizes this function, in part, by sequestering ß-catenin away from the pre-initiation complex. In conclusion, we present evidence that ß-catenin fulfills a previously unrecognized function in translational repression.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Peptide Chain Initiation, Translational , beta Catenin/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cycloheximide/pharmacology , Gene Ontology , HEK293 Cells , Humans , Mice , Peptide Chain Initiation, Translational/drug effects , Protein Binding/drug effects , Rats , Wnt Signaling Pathway/drug effects
15.
Nat Chem Biol ; 14(9): 844-852, 2018 09.
Article in English | MEDLINE | ID: mdl-29988067

ABSTRACT

Primordial germ cells (PGCs) form during early embryogenesis with a supply of maternal mRNAs that contain shorter poly(A) tails. How translation of maternal mRNAs is regulated during PGC development remains elusive. Here we describe a small-molecule screen with zebrafish embryos that identified primordazine, a compound that selectively ablates PGCs. Primordazine's effect on PGCs arises from translation repression through primordazine-response elements in the 3' UTRs. Systematic dissection of primordazine's mechanism of action revealed that translation of mRNAs during early embryogenesis occurs by two distinct pathways, depending on the length of their poly(A) tails. In addition to poly(A)-tail-dependent translation (PAT), early embryos perform poly(A)-tail-independent noncanonical translation (PAINT) via deadenylated 3' UTRs. Primordazine inhibits PAINT without inhibiting PAT, an effect that was also observed in quiescent, but not proliferating, mammalian cells. These studies reveal that PAINT is an alternative form of translation in the early embryo and is indispensable for PGC maintenance.


Subject(s)
3' Untranslated Regions/genetics , Germ Cells/metabolism , Peptide Chain Initiation, Translational/genetics , Animals , Cell Line, Tumor , Hydrazines/pharmacology , Mice , Peptide Chain Initiation, Translational/drug effects , Zebrafish
16.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2169-2182, 2018 06.
Article in English | MEDLINE | ID: mdl-29604334

ABSTRACT

Insulin resistance, defined as attenuated sensitivity responding to insulin, impairs insulin action. Direct causes and molecular mechanisms of insulin resistance have thus far remained elusive. Here we show that alternative translation initiation (ATI) of Caveolin-2 (Cav-2) regulates insulin sensitivity. Cav-2ß isoform yielded by ATI desensitizes insulin receptor (IR) via dephosphorylation by protein-tyrosine phosphatase 1B (PTP1B), and subsequent endocytosis and lysosomal degradation of IR, causing insulin resistance. Blockage of Cav-2 ATI protects against insulin resistance by preventing Cav-2ß-PTP1B-directed IR desensitization, thereby normalizing insulin sensitivity and glucose uptake. Our findings show that Cav-2ß is a negative regulator of IR signaling, and identify a mechanism causing insulin resistance through control of insulin sensitivity via Cav-2 ATI.


Subject(s)
Antigens, CD/metabolism , Caveolin 2/metabolism , Insulin Resistance/genetics , Peptide Chain Initiation, Translational/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Receptor, Insulin/metabolism , 3T3 Cells , Animals , Antigens, CD/genetics , Caveolin 2/genetics , Codon, Initiator/genetics , Endocytosis , HEK293 Cells , Humans , Lysosomes/metabolism , Mice , Mutagenesis, Site-Directed , Peptide Chain Initiation, Translational/drug effects , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Proteolysis , RNA Interference , RNA, Small Interfering/metabolism , Receptor, Insulin/genetics
17.
Exp Neurol ; 299(Pt B): 299-307, 2018 01.
Article in English | MEDLINE | ID: mdl-28610844

ABSTRACT

Meningiomas frequently display activation of the PI3K/AKT/mTOR pathway, leading to elevated levels of phospho-eukaryotic translation initiation factor 4E binding proteins, which enhances protein synthesis; however, it is not known whether inhibition of protein translation is an effective treatment option for meningiomas. We found that human meningiomas expressed high levels of the three components of the eukaryotic initiation factor 4F (eIF4F) translation initiation complex, eIF4A, eIF4E, and eIF4G. The expression of eIF4A and eIF4E was important in sustaining the growth of NF2-deficient benign meningioma Ben-Men-1 cells, as shRNA-mediated knockdown of these proteins strongly reduced cell proliferation. Among a series of 23 natural compounds evaluated, silvestrol, which inhibits eIF4A, was identified as being the most growth inhibitory in both primary meningioma and Ben-Men-1 cells. Silvestrol treatment of meningioma cells prominently induced G2/M arrest. Consistently, silvestrol significantly decreased the amounts of cyclins D1, E1, A, and B, PCNA, and Aurora A. In addition, total and phosphorylated AKT, ERK, and FAK, which have been shown to be important drivers for meningioma cell proliferation, were markedly lower in silvestrol-treated Ben-Men-1 cells. Our findings suggest that inhibiting protein translation could be a potential treatment for meningiomas.


Subject(s)
Antineoplastic Agents/pharmacology , Eukaryotic Initiation Factor-4A/biosynthesis , Eukaryotic Initiation Factor-4E/biosynthesis , Eukaryotic Initiation Factor-4G/biosynthesis , Meningeal Neoplasms/drug therapy , Meningioma/drug therapy , Neoplasm Proteins/biosynthesis , Peptide Chain Initiation, Translational/drug effects , Triterpenes/pharmacology , Antineoplastic Agents/therapeutic use , Aurora Kinase A/biosynthesis , Aurora Kinase A/genetics , Cyclins/biosynthesis , Cyclins/genetics , Drug Screening Assays, Antitumor , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4G/genetics , Female , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Male , Meningeal Neoplasms/genetics , Meningeal Neoplasms/pathology , Meningioma/genetics , Meningioma/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Proliferating Cell Nuclear Antigen/biosynthesis , Proliferating Cell Nuclear Antigen/genetics , RNA, Small Interfering/pharmacology , Triterpenes/therapeutic use , Tumor Cells, Cultured
18.
Mol Cell ; 68(3): 504-514.e7, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29107534

ABSTRACT

In eukaryotic cells, protein synthesis typically begins with the binding of eIF4F to the 7-methylguanylate (m7G) cap found on the 5' end of the majority of mRNAs. Surprisingly, overall translational output remains robust under eIF4F inhibition. The broad spectrum of eIF4F-resistant translatomes is incompatible with cap-independent translation mediated by internal ribosome entry sites (IRESs). Here, we report that N6-methyladenosine (m6A) facilitates mRNA translation that is resistant to eIF4F inactivation. Depletion of the methyltransferase METTL3 selectively inhibits translation of mRNAs bearing 5' UTR methylation, but not mRNAs with 5' terminal oligopyrimidine (TOP) elements. We identify ABCF1 as a critical mediator of m6A-promoted translation under both stress and physiological conditions. Supporting the role of ABCF1 in m6A-facilitated mRNA translation, ABCF1-sensitive transcripts largely overlap with METTL3-dependent mRNA targets. By illustrating the scope and mechanism of eIF4F-independent mRNA translation, these findings reshape our current perceptions of cellular translational pathways.


Subject(s)
Adenosine/analogs & derivatives , Eukaryotic Initiation Factor-4F/metabolism , Peptide Chain Initiation, Translational/drug effects , RNA Caps/genetics , RNA, Messenger/metabolism , 5' Untranslated Regions/genetics , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine/pharmacology , Eukaryotic Initiation Factor-4F/genetics , HeLa Cells , Humans , Internal Ribosome Entry Sites , Methyltransferases/genetics , Methyltransferases/metabolism , RNA Caps/drug effects , RNA, Messenger/genetics
19.
Methods Mol Biol ; 1520: 237-245, 2017.
Article in English | MEDLINE | ID: mdl-27873256

ABSTRACT

A strategy that can be applied to the research of new molecules with antibacterial activity is to look for inhibitors of essential bacterial processes within large collections of chemically heterogeneous compounds. The implementation of this approach requires the development of proper assays aimed at the identification of molecules interfering with specific cell pathways and potentially applicable to the high throughput analysis of large chemical library. Here, I describe a fluorescence-based whole-cell assay in Escherichia coli devised to find inhibitors of the translation initiation pathway. Translation is a complex and essential mechanism. It involves numerous sub-steps performed by factors that are in many cases sufficiently dissimilar in bacterial and eukaryotic cells to be targetable with domain-specific drugs. As a matter of fact, translation has been proven as one of the few bacterial mechanisms pharmacologically tractable with specific antibiotics. The assay described in this chapter is tailored to the identification of molecules affecting the first stage of translation initiation, which is the most dissimilar step in bacteria vs. mammals. The effect of the compounds under analysis is assayed in living cells, thus allowing evaluating their in vivo performance as inhibitors of translation initiation. Compared with other assays for antibacterials, the major advantages of this screen are its simplicity and high mechanism specificity.


Subject(s)
Biological Assay/methods , Peptide Chain Initiation, Translational/drug effects , Protein Synthesis Inhibitors/analysis , Protein Synthesis Inhibitors/pharmacology , Escherichia coli/cytology , Fluorescence
20.
Antiviral Res ; 137: 76-81, 2017 01.
Article in English | MEDLINE | ID: mdl-27864075

ABSTRACT

The DEAD-box RNA helicase eIF4A, which is part of the heterotrimeric translation initiation complex in eukaryotes, is an important novel drug target in cancer research because its helicase activity is required to unwind extended and highly structured 5'-UTRs of several proto-oncogenes. Silvestrol, a natural compound isolated from the plant Aglaia foveolata, is a highly efficient, non-toxic and specific inhibitor of eIF4A. Importantly, 5'-capped viral mRNAs often contain structured 5'-UTRs as well, which may suggest a dependence on eIF4A for their translation by the host protein synthesis machinery. In view of the recent Ebola virus (EBOV) outbreak in West Africa, the identification of potent antiviral compounds is urgently required. Since Ebola mRNAs are 5'-capped and harbor RNA secondary structures in their extended 5'-UTRs, we initiated a BSL4 study to analyze silvestrol in EBOV-infected Huh-7 cells and in primary human macrophages for its antiviral activity. We observed that silvestrol inhibits EBOV infection at low nanomolar concentrations, as inferred from large reductions of viral titers. This correlated with an almost complete disappearance of EBOV proteins, comparable in effect to the translational shutdown of expression of the proto-oncoprotein PIM1, a cellular kinase known to be affected by silvestrol. Effective silvestrol concentrations were non-toxic in the tested cell systems. Thus, silvestrol appears to be a promising first-line drug for the treatment of acute EBOV and possibly other viral infections.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Ebolavirus/drug effects , Macrophages/virology , Triterpenes/pharmacology , Virus Replication/drug effects , 5' Untranslated Regions/drug effects , Africa, Western/epidemiology , Antiviral Agents/toxicity , Cell Line, Tumor , Cells, Cultured , Ebolavirus/genetics , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/epidemiology , Humans , Macrophages/drug effects , Peptide Chain Initiation, Translational/drug effects , Proto-Oncogene Proteins c-pim-1/metabolism , RNA Caps/genetics , RNA Caps/metabolism , Triterpenes/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...