Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Nat Commun ; 12(1): 4217, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244513

ABSTRACT

The functional consequences of genetic variants within 5' untranslated regions (UTRs) on a genome-wide scale are poorly understood in disease. Here we develop a high-throughput multi-layer functional genomics method called PLUMAGE (Pooled full-length UTR Multiplex Assay on Gene Expression) to quantify the molecular consequences of somatic 5' UTR mutations in human prostate cancer. We show that 5' UTR mutations can control transcript levels and mRNA translation rates through the creation of DNA binding elements or RNA-based cis-regulatory motifs. We discover that point mutations can simultaneously impact transcript and translation levels of the same gene. We provide evidence that functional 5' UTR mutations in the MAP kinase signaling pathway can upregulate pathway-specific gene expression and are associated with clinical outcomes. Our study reveals the diverse mechanisms by which the mutational landscape of 5' UTRs can co-opt gene expression and demonstrates that single nucleotide alterations within 5' UTRs are functional in cancer.


Subject(s)
5' Untranslated Regions/genetics , DNA Mutational Analysis/methods , Gene Expression Regulation, Neoplastic , Genomics/methods , Prostatic Neoplasms/genetics , Cell Line, Tumor , HEK293 Cells , High-Throughput Screening Assays , Humans , Male , Point Mutation , Prostate/pathology , Prostatic Neoplasms/pathology , Protein Biosynthesis/genetics , RNA-Seq
3.
Curr Biol ; 31(1): 128-137.e5, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33157031

ABSTRACT

The translation machinery is composed of a myriad of proteins and RNAs whose levels must be coordinated to efficiently produce proteins without wasting energy or substrate. However, protein synthesis is clearly not always perfectly tuned to its environment, as disruption of translation machinery components can lengthen lifespan and stress survival. While much has been learned from bacteria and yeast about translational regulation, much less is known in metazoans. In a screen for mutations protecting C. elegans from hypoxic stress, we isolated multiple genes impacting protein synthesis: a ribosomal RNA helicase gene, tRNA biosynthesis genes, and a gene controlling amino acid availability. To define better the mechanisms by which these genes impact protein synthesis, we performed a second screen for suppressors of the conditional developmental arrest phenotype of the RNA helicase mutant and identified genes involved in ribosome biogenesis. Surprisingly, these suppressor mutations restored normal hypoxic sensitivity and protein synthesis to the tRNA biogenesis mutants, but not to the mutant reducing amino acid uptake. Proteomic analysis demonstrated that reduced tRNA biosynthetic activity produces a selective homeostatic reduction in ribosomal subunits, thereby offering a mechanism for the suppression results. Our study uncovers an unrecognized higher-order-translation regulatory mechanism in a metazoan whereby ribosome biogenesis genes communicate with genes controlling tRNA abundance matching the global rate of protein synthesis with available resources.


Subject(s)
Cell Hypoxia/genetics , Gene Expression Regulation , Protein Biosynthesis/genetics , RNA, Transfer/biosynthesis , Ribosomes/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Feedback, Physiological , Homeostasis/genetics , Mutation , RNA Helicases/genetics , RNA Helicases/metabolism
4.
Nat Commun ; 10(1): 4596, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601799

ABSTRACT

Many of the regulatory features governing erythrocyte specification, maturation, and associated disorders remain enigmatic. To identify new regulators of erythropoiesis, we utilize a functional genomic screen for genes affecting expression of the erythroid marker CD235a/GYPA. Among validating hits are genes coding for the N6-methyladenosine (m6A) mRNA methyltransferase (MTase) complex, including, METTL14, METTL3, and WTAP. We demonstrate that m6A MTase activity promotes erythroid gene expression programs through selective translation of ~300 m6A marked mRNAs, including those coding for SETD histone methyltransferases, ribosomal components, and polyA RNA binding proteins. Remarkably, loss of m6A marks results in dramatic loss of H3K4me3 marks across key erythroid-specific KLF1 transcriptional targets (e.g., Heme biosynthesis genes). Further, each m6A MTase subunit and a subset of their mRNAs targets are required for human erythroid specification in primary bone-marrow derived progenitors. Thus, m6A mRNA marks promote the translation of a network of genes required for human erythropoiesis.


Subject(s)
Adenosine/analogs & derivatives , Erythropoiesis/genetics , Protein Biosynthesis , Adenosine/genetics , Antigens, CD34/genetics , Antigens, CD34/metabolism , Bone Marrow Cells/physiology , CRISPR-Cas Systems , Cell Cycle Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukemia, Erythroblastic, Acute/genetics , Methyltransferases/genetics , Promoter Regions, Genetic , RNA Splicing Factors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulon
5.
Sci Transl Med ; 11(503)2019 07 31.
Article in English | MEDLINE | ID: mdl-31366581

ABSTRACT

The androgen receptor (AR) is a driver of cellular differentiation and prostate cancer development. An extensive body of work has linked these normal and aberrant cellular processes to mRNA transcription; however, the extent to which AR regulates posttranscriptional gene regulation remains unknown. Here, we demonstrate that AR uses the translation machinery to shape the cellular proteome. We show that AR is a negative regulator of protein synthesis and identify an unexpected relationship between AR and the process of translation initiation in vivo. This is mediated through direct transcriptional control of the translation inhibitor 4EBP1. We demonstrate that lowering AR abundance increases the assembly of the eIF4F translation initiation complex, which drives enhanced tumor cell proliferation. Furthermore, we uncover a network of pro-proliferation mRNAs characterized by a guanine-rich cis-regulatory element that is particularly sensitive to eIF4F hyperactivity. Using both genetic and pharmacologic methods, we demonstrate that dissociation of the eIF4F complex reverses the proliferation program, resulting in decreased tumor growth and improved survival in preclinical models. Our findings reveal a druggable nexus that functionally links the processes of mRNA transcription and translation initiation in an emerging class of lethal AR-deficient prostate cancer.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Regulon/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/genetics , Cell Proliferation/physiology , Humans , In Vitro Techniques , Introns/genetics , Male , Mice , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Regulon/genetics
6.
Oncogene ; 38(5): 687-698, 2019 01.
Article in English | MEDLINE | ID: mdl-30171262

ABSTRACT

Myelodysplastic syndromes (MDSs) are stem cell disorders with risk of transformation to acute myeloid leukemia (AML). Gene expression profiling reveals transcriptional expression of GLI1, of Hedgehog (Hh) signaling, in poor-risk MDS/AML. Using a murine model of MDS we demonstrated that constitutive Hh/Gli1 activation accelerated leukemic transformation and decreased overall survival. Hh/Gli1 activation resulted in clonal expansion of phenotypically defined granulocyte macrophage progenitors (GMPs) and acquisition of self-renewal potential in a non-self-renewing progenitor compartment. Transcriptome analysis of GMPs revealed enrichment in gene signatures of self-renewal pathways, operating via direct Gli1 activation. Using human cell lines we demonstrated that in addition to canonical Hh signaling, GLI1 is activated in a Smoothened-independent manner. GLI1 knockdown or inhibition with GANT61 resulted in decreased proliferation and clonogenic potential. Our data suggest that GLI1 activation is frequent in MDS during disease progression and inhibition of GLI1 is an attractive therapeutic target for a subset of patients.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Granulocyte-Macrophage Progenitor Cells/metabolism , Hedgehog Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Myelodysplastic Syndromes/metabolism , Zinc Finger Protein GLI1/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Granulocyte-Macrophage Progenitor Cells/pathology , Hedgehog Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Pyridines/pharmacology , Pyrimidines/pharmacology , Zinc Finger Protein GLI1/genetics
7.
Cell ; 173(5): 1204-1216.e26, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29628141

ABSTRACT

Pseudouridylation (Ψ) is the most abundant and widespread type of RNA epigenetic modification in living organisms; however, the biological role of Ψ remains poorly understood. Here, we show that a Ψ-driven posttranscriptional program steers translation control to impact stem cell commitment during early embryogenesis. Mechanistically, the Ψ "writer" PUS7 modifies and activates a novel network of tRNA-derived small fragments (tRFs) targeting the translation initiation complex. PUS7 inactivation in embryonic stem cells impairs tRF-mediated translation regulation, leading to increased protein biosynthesis and defective germ layer specification. Remarkably, dysregulation of this posttranscriptional regulatory circuitry impairs hematopoietic stem cell commitment and is common to aggressive subtypes of human myelodysplastic syndromes. Our findings unveil a critical function of Ψ in directing translation control in stem cells with important implications for development and disease.


Subject(s)
Intramolecular Transferases/metabolism , Protein Biosynthesis , Pseudouridine/metabolism , RNA, Transfer/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Differentiation , Eukaryotic Initiation Factors/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Intramolecular Transferases/antagonists & inhibitors , Intramolecular Transferases/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Myelodysplastic Syndromes/pathology , Nucleic Acid Conformation , Phosphoproteins/metabolism , Poly(A)-Binding Protein I/antagonists & inhibitors , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Protein I/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Stem Cell Niche
8.
Sci Transl Med ; 7(291): 291ra96, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26062848

ABSTRACT

FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations resulting in constitutive kinase activity are common in acute myeloid leukemia (AML) and carry a poor prognosis. Several agents targeting FLT3 have been developed, but their limited clinical activity suggests that the inhibition of other factors contributing to the malignant phenotype is required. We examined gene expression data sets as well as primary specimens and found that the expression of GLI2, a major effector of the Hedgehog (Hh) signaling pathway, was increased in FLT3-ITD compared to wild-type FLT3 AML. To examine the functional role of the Hh pathway, we studied mice in which Flt3-ITD expression results in an indolent myeloproliferative state and found that constitutive Hh signaling accelerated the development of AML by enhancing signal transducer and activator of transcription 5 (STAT5) signaling and the proliferation of bone marrow myeloid progenitors. Furthermore, combined FLT3 and Hh pathway inhibition limited leukemic growth in vitro and in vivo, and this approach may serve as a therapeutic strategy for FLT3-ITD AML.


Subject(s)
Hedgehog Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Mutant Proteins/metabolism , Signal Transduction , fms-Like Tyrosine Kinase 3/metabolism , Animals , Cell Compartmentation , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Progression , Drug Synergism , Gene Duplication/drug effects , Humans , Kruppel-Like Transcription Factors/metabolism , Mice , Myeloproliferative Disorders/pathology , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Nuclear Proteins/metabolism , Phenylurea Compounds/pharmacology , Receptors, G-Protein-Coupled/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Smoothened Receptor , Sorafenib , Stem Cells/cytology , Veratrum Alkaloids/pharmacology , Zinc Finger Protein Gli2
9.
Blood ; 123(5): 725-33, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24345755

ABSTRACT

Disease relapse remains a major factor limiting the survival of cancer patients. In the plasma cell malignancy multiple myeloma (MM), nearly all patients ultimately succumb to disease relapse and progression despite new therapies that have improved remission rates. Tumor regrowth indicates that clonogenic growth potential is continually maintained, but the determinants of self-renewal in MM are not well understood. Normal stem cells are regulated by extrinsic niche factors, and the tumor microenvironment (TME) may similarly influence tumor cell clonogenic growth and self-renewal. Growth differentiation factor 15 (GDF15) is aberrantly secreted by bone marrow stromal cells (BMSCs) in MM. We found that GDF15 is produced by BMSCs after direct contact with plasma cells and enhances the tumor-initiating potential and self-renewal of MM cells in a protein kinase B- and SRY (sex-determining region Y)-box-dependent manner. Moreover, GDF15 induces the expansion of MM tumor-initiating cells (TICs), and changes in the serum levels of GDF15 were associated with changes in the frequency of clonogenic MM cells and the progression-free survival of MM patients. These findings demonstrate that GDF15 plays a critical role in mediating the interaction among mature tumor cells, the TME, and TICs, and strategies targeting GDF15 may affect long-term clinical outcomes in MM.


Subject(s)
Growth Differentiation Factor 15/metabolism , Multiple Myeloma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Growth Differentiation Factor 15/blood , Humans , Mice , Mice, Inbred C57BL , Multiple Myeloma/blood , Multiple Myeloma/pathology , Proto-Oncogene Proteins c-akt/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction
10.
Int J Radiat Oncol Biol Phys ; 84(3): 800-6, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22445004

ABSTRACT

PURPOSE: We have previously shown that the antimalarial agent chloroquine can abrogate the lethal cellular effects of low-dose-rate (LDR) radiation in vitro, most likely by activating the ataxia-telangiectasia mutated (ATM) protein. Here, we demonstrate that chloroquine treatment also protects against lethal doses of LDR radiation in vivo. METHODS AND MATERIALS: C57BL/6 mice were irradiated with a total of 12.8 Gy delivered at 9.4 cGy/hour. ATM null mice from the same background were used to determine the influence of ATM. Chloroquine was administered by two intraperitoneal injections of 59.4 µg per 17 g of body weight, 24 hours and 4 hours before irradiation. Bone marrow cells isolated from tibia, fibula, and vertebral bones were transplanted into lethally irradiated CD45 congenic recipient mice by retroorbital injection. Chimerism was assessed by flow cytometry. In vitro methylcellulose colony-forming assay of whole bone marrow cells and fluorescence activated cell sorting analysis of lineage depleted cells were used to assess the effect of chloroquine on progenitor cells. RESULTS: Mice pretreated with chloroquine before radiation exhibited a significantly higher survival rate than did mice treated with radiation alone (80% vs. 31%, p = 0.0026). Chloroquine administration before radiation did not affect the survival of ATM null mice (p = 0.86). Chloroquine also had a significant effect on the early engraftment of bone marrow cells from the irradiated donor mice 6 weeks after transplantation (4.2% vs. 0.4%, p = 0.015). CONCLUSION: Chloroquine administration before radiation had a significant effect on the survival of normal but not ATM null mice, strongly suggesting that the in vivo effect, like the in vitro effect, is also ATM dependent. Chloroquine improved the early engraftment of bone marrow cells from LDR-irradiated mice, presumably by protecting the progenitor cells from radiation injury. Chloroquine thus could serve as a very useful drug for protection against the harmful effects of LDR radiation.


Subject(s)
Bone Marrow Cells/radiation effects , Cell Cycle Proteins/metabolism , Chloroquine/pharmacology , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/pharmacology , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Bone Marrow Transplantation/methods , Cell Cycle Proteins/genetics , Chloroquine/administration & dosage , DNA-Binding Proteins/genetics , Flow Cytometry/methods , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Radiation Chimera , Radiation Injuries, Experimental/mortality , Radiation-Protective Agents/administration & dosage , Survival Rate , Tumor Suppressor Proteins/genetics
11.
Crit Rev Eukaryot Gene Expr ; 20(2): 129-39, 2010.
Article in English | MEDLINE | ID: mdl-21133842

ABSTRACT

The Hedgehog signaling pathway is highly conserved and plays an essential role in the embryonic development of a wide variety of organs. In adult tissues, such as the central nervous system, it may also be required for homeostasis and repair following injury. The role of Hedgehog signaling in regulating hematopoiesis is not entirely clear. Evidence has shown that Hedgehog signaling is required for both primitive hematopoiesis in the developing embryo, as well as for definitive hematopoiesis in the adult. However, several studies also suggest that Hedgehog pathway activity is completely dispensable in postnatal hematopoiesis. In this review, we discuss the current understanding of Hedgehog signaling in vertebrate hematopoiesis, as well as the contradictory findings that have been reported.


Subject(s)
Hedgehog Proteins/metabolism , Hematopoiesis/physiology , Signal Transduction , Animals , Humans
12.
Protein Sci ; 15(1): 182-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16322573

ABSTRACT

Despite the availability of numerous gene fusion systems, recombinant protein expression in Escherichia coli remains difficult. Establishing the best fusion partner for difficult-to-express proteins remains empirical. To determine which fusion tags are best suited for difficult-to-express proteins, a comparative analysis of the newly described SUMO fusion system with a variety of commonly used fusion systems was completed. For this study, three model proteins, enhanced green fluorescent protein (eGFP), matrix metalloprotease-13 (MMP13), and myostatin (growth differentiating factor-8, GDF8), were fused to the C termini of maltose-binding protein (MBP), glutathione S-transferase (GST), thioredoxin (TRX), NUS A, ubiquitin (Ub), and SUMO tags. These constructs were expressed in E. coli and evaluated for expression and solubility. As expected, the fusion tags varied in their ability to produce tractable quantities of soluble eGFP, MMP13, and GDF8. SUMO and NUS A fusions enhanced expression and solubility of recombinant proteins most dramatically. The ease at which SUMO and NUS A fusion tags were removed from their partner proteins was then determined. SUMO fusions are cleaved by the natural SUMO protease, while an AcTEV protease site had to be engineered between NUS A and its partner protein. A kinetic analysis showed that the SUMO and AcTEV proteases had similar KM values, but SUMO protease had a 25-fold higher kcat than AcTEV protease, indicating a more catalytically efficient enzyme. Taken together, these results demonstrate that SUMO is superior to commonly used fusion tags in enhancing expression and solubility with the distinction of generating recombinant protein with native sequences.


Subject(s)
Cloning, Molecular/methods , Gene Fusion , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , SUMO-1 Protein/biosynthesis , SUMO-1 Protein/genetics , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Endopeptidases/biosynthesis , Endopeptidases/chemistry , Endopeptidases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Recombinant Fusion Proteins/chemistry , SUMO-1 Protein/chemistry , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...