Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Ther ; 29(3): 1164-1173, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33186690

ABSTRACT

Synthetic mRNA therapeutics have the potential to revolutionize healthcare, as they enable patients to produce therapeutic proteins inside their own bodies. However, convenient methods that allow external control over the timing and magnitude of protein production after in vivo delivery of synthetic mRNA are lacking. In this study, we validate the in vivo utility of a synthetic self-amplifying mRNA (RNA replicon) whose expression can be turned off using a genetic switch that responds to oral administration of trimethoprim (TMP), a US Food and Drug Administration (FDA)-approved small-molecule drug. After intramuscular electroporation, the engineered RNA replicon exhibited dose-dependent and reversible expression of its encoded protein upon TMP administration. The TMP serum level needed for maximal downregulation of protein translation was approximately 45-fold below that used in humans for therapeutic purposes. To demonstrate the therapeutic potential of the technology, we injected mice with a TMP-responsive RNA replicon encoding erythropoietin (EPO) and successfully controlled the timing and magnitude of EPO production as well as changes in hematocrit. This work demonstrates the feasibility of controlling mRNA kinetics in vivo, thereby broadly expanding the clinical versatility of mRNA therapeutics.


Subject(s)
Erythropoietin/metabolism , Folic Acid Antagonists/administration & dosage , Protein Biosynthesis , RNA, Messenger/metabolism , Replicon , Trimethoprim/administration & dosage , Animals , Electroporation , Erythropoietin/genetics , Female , Genetic Therapy , Injections, Intramuscular , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics
2.
Nat Chem Biol ; 14(11): 1043-1050, 2018 11.
Article in English | MEDLINE | ID: mdl-30327560

ABSTRACT

Synthetic mRNA is an attractive vehicle for gene therapies because of its transient nature and improved safety profile over DNA. However, unlike DNA, broadly applicable methods to control expression from mRNA are lacking. Here we describe a platform for small-molecule-based regulation of expression from modified RNA (modRNA) and self-replicating RNA (replicon) delivered to mammalian cells. Specifically, we engineer small-molecule-responsive RNA binding proteins to control expression of proteins from RNA-encoded genetic circuits. Coupled with specific modRNA dosages or engineered elements from a replicon, including a subgenomic promoter library, we demonstrate the capability to externally regulate the timing and level of protein expression. These control mechanisms facilitate the construction of ON, OFF, and two-output switches, with potential therapeutic applications such as inducible cancer immunotherapies. These circuits, along with other synthetic networks that can be developed using these tools, will expand the utility of synthetic mRNA as a therapeutic modality.


Subject(s)
Gene Regulatory Networks , Genetic Therapy/methods , Promoter Regions, Genetic , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , RNA/chemistry , Animals , Cell Line , Cricetinae , DNA/chemistry , Gene Library , Genetic Engineering , HEK293 Cells , Humans , Immunotherapy , Mice , RNA, Small Interfering/metabolism , Synthetic Biology
3.
Science ; 359(6376)2018 02 09.
Article in English | MEDLINE | ID: mdl-29439214

ABSTRACT

Gene and engineered-cell therapies promise to treat diseases by genetically modifying cells to carry out therapeutic tasks. Although the field has had some success in treating monogenic disorders and hematological malignancies, current approaches are limited to overexpression of one or a few transgenes, constraining the diseases that can be treated with this approach and leading to potential concerns over safety and efficacy. Synthetic gene networks can regulate the dosage, timing, and localization of gene expression and therapeutic activity in response to small molecules and disease biomarkers. Such "programmable" gene and engineered-cell therapies will provide new interventions for incurable or difficult-to-treat diseases.


Subject(s)
Cell Engineering/methods , Cell- and Tissue-Based Therapy , Cellular Reprogramming Techniques , Genetic Engineering/methods , Genetic Therapy , Synthetic Biology/methods , DNA/genetics , Gene Expression , Gene Regulatory Networks , Genes, Synthetic , Humans , RNA/genetics , Recombinant Fusion Proteins , Transgenes
4.
J Control Release ; 217: 337-44, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26342664

ABSTRACT

Messenger RNA as a therapeutic modality is becoming increasingly popular in the field of gene therapy. The realization that nucleobase modifications can greatly enhance the properties of mRNA by reducing the immunogenicity and increasing the stability of the RNA molecule (the Kariko paradigm) has been pivotal for this revolution. Here we find that mRNAs containing the N(1)-methylpseudouridine (m1Ψ) modification alone and/or in combination with 5-methylcytidine (m5C) outperformed the current state-of-the-art pseudouridine (Ψ) and/or m5C/Ψ-modified mRNA platform by providing up to ~44-fold (when comparing double modified mRNAs) or ~13-fold (when comparing single modified mRNAs) higher reporter gene expression upon transfection into cell lines or mice, respectively. We show that (m5C/)m1Ψ-modified mRNA resulted in reduced intracellular innate immunogenicity and improved cellular viability compared to (m5C/)Ψ-modified mRNA upon in vitro transfection. The enhanced capability of (m5C/)m1Ψ-modified mRNA to express proteins may at least partially be due to the increased ability of the mRNA to evade activation of endosomal Toll-like receptor 3 (TLR3) and downstream innate immune signaling. We believe that the (m5C/)m1Ψ-mRNA platform presented here may serve as a new standard in the field of modified mRNA-based therapeutics.


Subject(s)
Cytidine/analogs & derivatives , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , RNA, Messenger/chemistry , Animals , Cell Line , Cell Line, Tumor , Cell Survival , Cytidine/chemistry , Humans , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Mice, Inbred BALB C , RNA, Messenger/pharmacology , Transfection
5.
Nat Biotechnol ; 33(8): 839-41, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26237515

ABSTRACT

Synthetic regulatory circuits encoded in RNA rather than DNA could provide a means to control cell behavior while avoiding potentially harmful genomic integration in therapeutic applications. We create post-transcriptional circuits using RNA-binding proteins, which can be wired in a plug-and-play fashion to create networks of higher complexity. We show that the circuits function in mammalian cells when encoded in modified mRNA or self-replicating RNA.


Subject(s)
Gene Regulatory Networks/genetics , RNA-Binding Proteins/genetics , RNA/genetics , Synthetic Biology/methods , Animals , Cell Line , Cricetinae , HEK293 Cells , Humans , RNA/metabolism , RNA-Binding Proteins/metabolism
6.
Genes Dev ; 29(4): 350-5, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25691465

ABSTRACT

Here we show that the Ino80 chromatin remodeling complex (Ino80C) directly prevents euchromatin from invading transcriptionally silent chromatin within intergenic regions and at the border of euchromatin and heterochromatin. Deletion of Ino80C subunits leads to increased H3K79 methylation and noncoding RNA polymerase II (Pol II) transcription centered at the Ino80C-binding sites. The effect of Ino80C is direct, as it blocks H3K79 methylation by Dot1 in vitro. Heterochromatin stimulates the binding of Ino80C in vitro and in vivo. Our data reveal that Ino80C serves as a general silencing complex that restricts transcription to gene units in euchromatin.


Subject(s)
Chromatin/genetics , Euchromatin/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Binding Sites , Euchromatin/genetics , Gene Expression Regulation, Fungal , Gene Silencing , Histone-Lysine N-Methyltransferase/metabolism , Methylation , Nuclear Proteins/metabolism , Protein Binding , RNA Polymerase II/metabolism
7.
Expert Rev Vaccines ; 14(2): 313-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25566800

ABSTRACT

Nucleic acid vaccines have been gaining attention as an alternative to the standard attenuated pathogen or protein based vaccine. However, an unrealized advantage of using such DNA or RNA based vaccination modalities is the ability to program within these nucleic acids regulatory devices that would provide an immunologist with the power to control the production of antigens and adjuvants in a desirable manner by administering small molecule drugs as chemical triggers. Advances in synthetic biology have resulted in the creation of highly predictable and modular genetic parts and devices that can be composed into synthetic gene circuits with complex behaviors. With the recent advent of modified RNA gene delivery methods and developments in the RNA replicon platform, we foresee a future in which mammalian synthetic biologists will create genetic circuits encoded exclusively on RNA. Here, we review the current repertoire of devices used in RNA synthetic biology and propose how programmable 'smart vaccines' will revolutionize the field of RNA vaccination.


Subject(s)
DNA/immunology , Proteins/immunology , RNA/immunology , Vaccination , Vaccines, DNA/immunology , DNA/genetics , Gene Regulatory Networks , Genetic Engineering , Genetic Therapy , Humans , Proteins/genetics , RNA/genetics , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , Synthetic Biology , Vaccines, DNA/genetics
8.
ACS Synth Biol ; 4(1): 48-56, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-24877739

ABSTRACT

RNA replicons are an emerging platform for engineering synthetic biological systems. Replicons self-amplify, can provide persistent high-level expression of proteins even from a small initial dose, and, unlike DNA vectors, pose minimal risk of chromosomal integration. However, no quantitative model sufficient for engineering levels of protein expression from such replicon systems currently exists. Here, we aim to enable the engineering of multigene expression from more than one species of replicon by creating a computational model based on our experimental observations of the expression dynamics in single- and multireplicon systems. To this end, we studied fluorescent protein expression in baby hamster kidney (BHK-21) cells using a replicon derived from Sindbis virus (SINV). We characterized expression dynamics for this platform based on the dose-response of a single species of replicon over 50 h and on a titration of two cotransfected replicons expressing different fluorescent proteins. From this data, we derive a quantitative model of multireplicon expression and validate it by designing a variety of three-replicon systems, with profiles that match desired expression levels. We achieved a mean error of 1.7-fold on a 1000-fold range, thus demonstrating how our model can be applied to precisely control expression levels of each Sindbis replicon species in a system.


Subject(s)
Genetic Engineering/methods , Models, Genetic , Animals , Cell Line , Cricetinae , Gene Expression , Luminescent Proteins/genetics , RNA, Viral/genetics , Replicon , Sindbis Virus/genetics , Synthetic Biology , Transfection
9.
Genes Dev ; 26(21): 2443-55, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23124068

ABSTRACT

Yeast contains heterochromatin at telomeres and the silent mating-type loci (HML/HMR). Genes positioned within the telomeric heterochromatin of Saccharomyces cerevisiae switch stochastically between epigenetically bistable ON and OFF expression states. Important aspects of the mechanism of variegated gene expression, including the chromatin structure of the natural ON state and the mechanism by which it is maintained, are unknown. To address this issue, we developed approaches to select cells in the ON and OFF states. We found by chromatin immunoprecipitation (ChIP) that natural ON telomeres are associated with Rap1 binding and, surprisingly, also contain known characteristics of OFF telomeres, including significant amounts of Sir3 and H4K16 deacetylated nucleosomes. Moreover, we found that H3K79 methylation (H3K79me), H3K4me, and H3K36me, which are depleted from OFF telomeres, are enriched at ON telomeres. We demonstrate in vitro that H3K79me, but not H3K4me or H3K36me, disrupts transcriptional silencing. Importantly, H3K79me does not significantly reduce Sir complex binding in vivo or in vitro. Finally, we show that maintenance of H3K79me at ON telomeres is dependent on transcription. Therefore, although Sir proteins are required for silencing, we propose that epigenetic variegation of telomeric gene expression is due to the bistable enrichment/depletion of H3K79me and not the fluctuation in the amount of Sir protein binding to nucleosomes.


Subject(s)
Epigenomics , Gene Expression Regulation, Fungal , Genetic Variation , Heterochromatin/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/genetics , Culture Media , DNA Methylation , Flow Cytometry , Gene Silencing , Histones/metabolism , Protein Binding , Saccharomyces cerevisiae Proteins/metabolism
10.
Curr Biol ; 22(1): 56-63, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22177115

ABSTRACT

The S. cerevisiae Rpd3 large (Rpd3L) and small (Rpd3S) histone deacetylase (HDAC) complexes are prototypes for understanding transcriptional repression in eukaryotes [1]. The current view is that they function by deacetylating chromatin, thereby limiting accessibility of transcriptional factors to the underlying DNA. However, an Rpd3 catalytic mutant retains substantial repression capability when targeted to a promoter as a LexA fusion protein [2]. We investigated the HDAC-independent properties of the Rpd3 complexes biochemically and discovered a chaperone function, which promotes histone deposition onto DNA, and a novel activity, which prevents nucleosome eviction but not remodeling mediated by the ATP-dependent RSC complex. These HDAC-independent activities inhibit Pol II transcription on a nucleosomal template. The functions of the endogenous Rpd3 complexes can be recapitulated with recombinant Rpd3 core complex comprising Sin3, Rpd3, and Ume1. To test the hypothesis that Rpd3 contributes to chromatin stabilization in vivo, we measured histone H3 density genomewide and found that it was reduced at promoters in an Rpd3 deletion mutant but partially restored in a catalytic mutant. Importantly, the effects on H3 density are most apparent on RSC-enriched genes [3]. Our data suggest that the Rpd3 core complex could contribute to repression via a novel nucleosome stabilization function.


Subject(s)
Chromatin/metabolism , Histone Deacetylases/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphate/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Fungal , Genome, Fungal , Histone Deacetylases/genetics , Histones/genetics , Histones/metabolism , Molecular Chaperones/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Proc Natl Acad Sci U S A ; 108(31): 12693-8, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21771901

ABSTRACT

DNA topoisomerases are believed to promote transcription by removing excessive DNA supercoils produced during elongation. However, it is unclear how topoisomerases in eukaryotes are recruited and function in the transcription pathway in the context of nucleosomes. To address this problem we present high-resolution genome-wide maps of one of the major eukaryotic topoisomerases, Topoisomerase II (Top2) and nucleosomes in the budding yeast, Saccharomyces cerevisiae. Our data indicate that at promoters Top2 binds primarily to DNA that is nucleosome-free. However, although nucleosome loss enables Top2 occupancy, the opposite is not the case and the loss of Top2 has little effect on nucleosome density. We also find that Top2 is involved in transcription. Not only is Top2 enriched at highly transcribed genes, but Top2 is required redundantly with Top1 for optimal recruitment of RNA polymerase II at their promoters. These findings and the examination of candidate-activated genes suggest that nucleosome loss induced by nucleosome remodeling factors during gene activation enables Top2 binding, which in turn acts redundantly with Top1 to enhance recruitment of RNA polymerase II.


Subject(s)
DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type I/metabolism , DNA, Fungal/metabolism , RNA Polymerase II/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Chromatin Immunoprecipitation , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type II/genetics , DNA, Fungal/genetics , Genome, Fungal/genetics , Mutation , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Proton-Phosphate Symporters/genetics , Proton-Phosphate Symporters/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription, Genetic
12.
Cell Cycle ; 10(2): 293-300, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21212735

ABSTRACT

Histones of heterochromatin are deacetylated in yeast and methylated in more complex eukaryotes to regulate heterochromatin structure and gene silencing. Here, we report that histone H2A phosphorylated at serine 129 (γH2A) in Saccharomyces cerevisiae is a conceptually new type of heterochromatin modification that functions downstream of silent chromatin assembly. We show that γH2A is enriched throughout yeast telomeric and silent mating locus (HM) heterochromatin where γH2A results from the action of kinases Tel1 and Mec1. Interestingly, mutation of γH2A has no apparent effect on the binding of Sir (silent information regulator) complex or on gene silencing. In contrast, deletion of SIR3 abolishes the formation of γH2A at heterochromatin. To address the function of γH2A, we used a Δrif1 mutant strain in which telomeres are excessively elongated to show that γH2A is required for the optimal recruitment of Cdc13, a regulator of telomere elongation, and for telomere elongation itself. Thus, a histone modification that parallels Sir3 protein binding is shown here to be dispensable for the formation of a silent structure but is important for a crucial heterochromatin-specific downstream function in telomere homeostasis.


Subject(s)
Heterochromatin/metabolism , Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Telomere/metabolism , Cell Division , Chromatin Immunoprecipitation , Gene Silencing , Histones/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mutation , Protein Serine-Threonine Kinases/metabolism , S Phase , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Telomere/physiology , Telomere-Binding Proteins/metabolism
13.
Mol Cell ; 32(3): 449-55, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18995842

ABSTRACT

Autoacetylation of the p300 histone acetyltransferase controls the transition between VP16-mediated chromatin acetylation and preinitiation complex (PIC) assembly. Currently, it is unknown if and how autoacetylated p300 is deacetylated. We found that the NAD(+)-dependent histone deacetylase SIRT2 deacetylates p300 in vitro and in cells. SIRT2 deacetylates lysine residues in the catalytic domain of p300 and restores binding of p300 to the PIC. RNAi-mediated depletion or chemical inhibition of SIRT2 in cells results in accumulation of acetylated p300. The altered ac-p300/p300 ratio in SIRT2-depleted cells results in decreased p300 recruitment to an integrated VP16-responsive gene and inhibition of transcription. We conclude that p300 undergoes a dynamic cycle of autoacetylation and deacetylation.


Subject(s)
Sirtuins/physiology , p300-CBP Transcription Factors/metabolism , Acetylation , Cell Nucleus/enzymology , Cytoplasm/enzymology , HeLa Cells , Homeostasis , Humans , Kinetics , Protein Processing, Post-Translational , Recombinant Proteins/metabolism , Sirtuin 2 , Sirtuins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...