Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
NPJ Vaccines ; 8(1): 157, 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37828126

ABSTRACT

Annually, seasonal influenza is responsible for millions of infections and hundreds of thousands of deaths. The current method for managing influenza is vaccination using a standardized amount of the influenza virus' primary surface antigen, hemagglutinin (HA), as the intended target of the immune response. This vaccination strategy results in vaccines with variable efficacy year to year due to antigenic drift of HA, which can be further exacerbated by manufacturing processes optimizing growth of vaccine virus in eggs. Due to these limitations, alternative vaccine platforms are actively being explored to improve influenza vaccine efficacy, including cell-based, recombinant protein, and mRNA vaccines. mRNA's rapid, in vitro production makes it an appealing platform for influenza vaccination, and the success of SARS-CoV-2 mRNA vaccines in the clinic has encouraged the development of mRNA vaccines for other pathogens. Here, the immunogenicity and protective efficacy of a quadrivalent mRNA vaccine encoding HA from four seasonal influenza viruses, A/California/07/2009 (H1N1), A/Hong Kong/4801/2014 (H3N2), B/Brisbane/60/2008 (B-Victoria lineage), and B/Phuket/3073/2013 (B-Yamagata lineage), was evaluated. In mice, a 120 µg total dose of this quadrivalent mRNA vaccine induced robust antibody titers against each subtype that were commensurate with titers when each antigen was administered alone. Following A/California/04/2009 challenge, mice were fully protected from morbidity and mortality, even at doses as low as 1 µg of each antigen. Additionally, a single administration of 10 µg of quadrivalent mRNA was sufficient to prevent weight loss caused by A/California/04/2009. These results support the promise of this mRNA vaccine for prevention and mitigation of influenza vaccine.

2.
Nat Commun ; 13(1): 1323, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35260576

ABSTRACT

Secreted proteins, such as hormones or cytokines, are key mediators in multicellular organisms. Response of protein secretion based on transcriptional control is rather slow, as it requires transcription, translation and transport from the endoplasmic reticulum (ER) to the plasma membrane via the conventional protein secretion (CPS) pathway. An alternative regulation to provide faster response would be valuable. Here we present two genetically encoded orthogonal regulatory secretion systems, which rely on the retention of pre-synthesized proteins on the ER membrane (membER, released by a cytosolic protease) or inside the ER lumen (lumER, released by an ER-luminal protease), respectively, and their release by the chemical signal-regulated proteolytic removal of an ER-retention signal, without triggering ER stress due to protein aggregates. Design of orthogonal chemically-regulated split proteases enables the combination of signals into logic functions. Its application was demonstrated on a chemically regulated therapeutic protein secretion and regulated membrane translocation of a chimeric antigen receptor (CAR) targeting cancer antigen. Regulation of the ER escape represents a platform for the design of fast-responsive and tightly-controlled modular and scalable protein secretion system for mammalian cells.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Animals , Endopeptidases/metabolism , Endoplasmic Reticulum/metabolism , Mammals/metabolism , Peptide Hydrolases/metabolism , Protein Transport , Proteolysis
3.
Nat Chem Biol ; 15(2): 115-122, 2019 02.
Article in English | MEDLINE | ID: mdl-30531965

ABSTRACT

Cellular signal transduction is predominantly based on protein interactions and their post-translational modifications, which enable a fast response to input signals. Owing to difficulties in designing new unique protein-protein interactions, designed cellular logic has focused on transcriptional regulation; however, that process has a substantially slower response, because it requires transcription and translation. Here, we present de novo design of modular, scalable signaling pathways based on proteolysis and designed coiled coils (CC) and implemented in mammalian cells. A set of split proteases with highly specific orthogonal cleavage motifs was constructed and combined with strategically positioned cleavage sites and designed orthogonal CC dimerizing domains with tunable affinity for competitive displacement after proteolytic cleavage. This framework enabled the implementation of Boolean logic functions and signaling cascades in mammalian cells. The designed split-protease-cleavable orthogonal-CC-based (SPOC) logic circuits enable response to chemical or biological signals within minutes rather than hours and should be useful for diverse medical and nonmedical applications.


Subject(s)
Protein Engineering/methods , Protein Interaction Mapping/methods , Animals , Endopeptidases , Gene Expression Regulation/genetics , Humans , Logic , Mammals , Protein Domains/physiology , Protein Processing, Post-Translational/physiology , Proteolysis , Signal Transduction , Synthetic Biology/methods
4.
Nucleic Acids Res ; 44(3): 1471-81, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26748097

ABSTRACT

Development of orthogonal, designable and adjustable transcriptional regulators is an important goal of synthetic biology. Their activity has been typically modulated through stimulus-induced oligomerization or interaction between the DNA-binding and activation/repression domain. We exploited a feature of the designable Transcription activator-like effector (TALE) DNA-binding domain that it winds around the DNA which allows to topologically prevent it from binding by intramolecular cyclization. This new approach was investigated through noncovalent ligand-induced cyclization or through a covalent split intein cyclization strategy, where the topological inhibition of DNA binding by cyclization and its restoration by a proteolytic release of the topologic constraint was expected. We show that locked TALEs indeed have diminished DNA binding and regain full transcriptional activity by stimulation with the rapamycin ligand or site-specific proteolysis of the peptide linker, with much higher level of activation than rapamycin-induced heterodimerization. Additionally, we demonstrated reversibility, activation of genomic targets and implemented logic gates based on combinations of protein cyclization, proteolytic cleavage and ligand-induced dimerization, where the strongest fold induction was achieved by the proteolytic cleavage of a repression domain from a linear TALE.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Trans-Activators/metabolism , Transcriptional Activation , Amino Acid Sequence , DNA/chemistry , DNA/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Models, Genetic , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Proteolysis , Trans-Activators/chemistry , Trans-Activators/genetics
5.
Nat Commun ; 5: 5007, 2014 Sep 29.
Article in English | MEDLINE | ID: mdl-25264186

ABSTRACT

Bistable switches are fundamental regulatory elements of complex systems, ranging from electronics to living cells. Designed genetic toggle switches have been constructed from pairs of natural transcriptional repressors wired to inhibit one another. The complexity of the engineered regulatory circuits can be increased using orthogonal transcriptional regulators based on designed DNA-binding domains. However, a mutual repressor-based toggle switch comprising DNA-binding domains of transcription-activator-like effectors (TALEs) did not support bistability in mammalian cells. Here, the challenge of engineering a bistable switch based on monomeric DNA-binding domains is solved via the introduction of a positive feedback loop composed of activators based on the same TALE domains as their opposing repressors and competition for the same DNA operator site. This design introduces nonlinearity and results in epigenetic bistability. This principle could be used to employ other monomeric DNA-binding domains such as CRISPR for applications ranging from reprogramming cells to building digital biological memory.


Subject(s)
DNA/chemistry , Genetic Engineering/methods , Binding Sites , Binding, Competitive , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Epigenesis, Genetic , HEK293 Cells , Humans , Luciferases/metabolism , Microscopy, Confocal , Models, Theoretical , Protein Binding , Protein Structure, Tertiary , Stochastic Processes
SELECTION OF CITATIONS
SEARCH DETAIL
...