Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Proc Natl Acad Sci U S A ; 111(5): E554-62, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449891

ABSTRACT

Aptazymes are small, ligand-dependent self-cleaving ribozymes that function independently of transcription factors and can be customized for induction by various small molecules. Here, we introduce these artificial riboswitches for regulation of DNA and RNA viruses. We hypothesize that they represent universally applicable tools for studying viral gene functions and for applications as a safety switch for oncolytic and live vaccine viruses. Our study shows that the insertion of artificial aptazymes into the adenoviral immediate early gene E1A enables small-molecule-triggered, dose-dependent inhibition of gene expression. Aptazyme-mediated shutdown of E1A expression translates into inhibition of adenoviral genome replication, infectious particle production, and cytotoxicity/oncolysis. These results provide proof of concept for the aptazyme approach for effective control of biological outcomes in eukaryotic systems, specifically in virus infections. Importantly, we also demonstrate aptazyme-dependent regulation of measles virus fusion protein expression, translating into potent reduction of progeny infectivity and virus spread. This not only establishes functionality of aptazymes in fully cytoplasmic genetic systems, but also implicates general feasibility of this strategy for application in viruses with either DNA or RNA genomes. Our study implies that gene regulation by artificial riboswitches may be an appealing alternative to Tet- and other protein-dependent gene regulation systems, based on their small size, RNA-intrinsic mode of action, and flexibility of the inducing molecule. Future applications range from gene analysis in basic research to medicine, for example as a safety switch for new generations of efficiency-enhanced oncolytic viruses.


Subject(s)
DNA Viruses/genetics , DNA Viruses/physiology , Gene Expression Regulation, Viral , RNA Viruses/genetics , RNA Viruses/physiology , Riboswitch/genetics , Virus Replication/genetics , Adenoviridae/genetics , Adenoviridae/pathogenicity , Adenoviridae/physiology , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Cell Line , DNA Viruses/pathogenicity , Genes, Viral/genetics , Ligands , Measles virus/genetics , Measles virus/pathogenicity , Measles virus/physiology , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , RNA Viruses/pathogenicity , RNA, Catalytic/metabolism , Virion/physiology , Virus Internalization
2.
Nucleic Acids Res ; 40(21): e167, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22885302

ABSTRACT

Therapeutic gene transfer by replication-defective viral vectors or, for cancer treatment, by replication-competent oncolytic viruses shows high promise for treatment of major diseases. To ensure safety, timing or dosing in patients, external control of therapeutic gene expression is desirable or even required. In this study, we explored the potential of artificial aptazymes, ligand-dependent self-cleaving ribozymes, as an innovative tool for regulation of therapeutic gene expression. Importantly, aptazymes act on RNA intrinsically, independent of regulatory protein-nucleic acid interactions and stoichiometry, are non-immunogenic and of small size. These are key advantages compared with the widely used inducible promoters, which were also reported to lose regulation at high copy numbers, e.g. after replication of oncolytic viruses. We characterized aptazymes in therapeutic gene transfer utilizing adenovectors (AdVs), adeno-associated vectors (AAVs) and oncolytic adenoviruses (OAds), which are all in advanced clinical testing. Our results show similar aptazyme-mediated regulation of gene expression by plasmids, AdVs, AAVs and OAds. Insertion into the 5'-, 3'- or both untranslated regions of several transgenes resulted in ligand-responsive gene expression. Notably, aptazyme regulation was retained during OAd replication and spread. In conclusion, our study demonstrates the fidelity of aptazymes in viral vectors and oncolytic viruses and highlights the potency of riboswitches for medical applications.


Subject(s)
Adenoviridae/genetics , Gene Expression Regulation , Oncolytic Viruses/genetics , RNA, Catalytic/genetics , Riboswitch , Adenoviridae/physiology , Cell Line, Tumor , Defective Viruses/genetics , Dependovirus/genetics , Genetic Vectors , Genome, Viral , Humans , RNA, Catalytic/metabolism , Transduction, Genetic , Transgenes , Untranslated Regions , Virus Replication
3.
PLoS One ; 9(4): e95723, 2014.
Article in English | MEDLINE | ID: mdl-24760010

ABSTRACT

Adenoviral gene therapy and oncolysis would critically benefit from targeted cell entry by genetically modified capsids. This requires both the ablation of native adenovirus tropism and the identification of ligands that remain functional in virus context. Here, we establish cell type-specific entry of HAdV-5-based vectors by genetic ligand insertion into a chimeric fiber with shaft and knob domains of the short HAdV-41 fiber (Ad5T/41sSK). This fiber format was reported to ablate transduction in vitro and biodistribution to the liver in vivo. We show that the YSA peptide, binding to the pan-cancer marker EphA2, can be inserted into three positions of the chimeric fiber, resulting in strong transduction of EphA2-positive but not EphA2-negative cells of human melanoma biopsies and of tumor xenografts after intratumoral injection. Transduction was blocked by soluble YSA peptide and restored for EphA2-negative cells after recombinant EphA2 expression. The YSA peptide could also be inserted into three positions of a CAR binding-ablated HAdV-5 fiber enabling specific transduction; however, the Ad5T/41sSK format was superior in vivo. In conclusion, we establish an adenovirus capsid facilitating functional insertion of targeting peptides and a novel adenovirus using the tumor marker EphA2 as receptor with high potential for cancer gene therapy and viral oncolysis.


Subject(s)
Adenoviridae/metabolism , Receptor, EphA2/metabolism , Animals , Cell Line , Female , Humans , Melanoma/metabolism , Melanoma/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays
5.
Mol Biosyst ; 6(5): 807-14, 2010 May.
Article in English | MEDLINE | ID: mdl-20567766

ABSTRACT

The possibility to externally control gene expression is of fundamental importance in both basic and applied life sciences. Although there are some techniques available to regulate expression in mammalian cells, they rely on the presence of ligand-sensing transcription factors, making it necessary to generate cell lines or organisms that stably express these regulatory factors. In recent years, mechanisms relying on direct RNA-ligand interactions for controlling gene expression have been both discovered in nature and engineered artificially. Among the latter, RNA switches relying on catalytically active RNA have been described. In principle, ligand-dependent triggering of mRNA self-cleavage should be a universal mechanism in order to control gene expression in a variety of organisms. Nevertheless, no examples of such aptazymes acting as RNA-based switches have been reported so far in mammalian cells. Here we present the theophylline-induced activation of an mRNA-based hammerhead ribozyme, resulting in an off-switch of gene expression. Starting from an artificial aptazyme switch reported to function in bacteria, we identified and optimized important parameters such as artificial start codons and the communicating sequence connecting ribozyme and aptamer, resulting in an RNA switch that allows for controlling transgenic expression in mammalian cells without the need to express a corresponding ligand-sensing transcription factor.


Subject(s)
Gene Expression Regulation , RNA, Catalytic/chemistry , RNA, Catalytic/metabolism , Base Sequence , Cell Line , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Models, Biological , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Catalytic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL