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










Publication year range
1.
Biotechnol J ; 17(7): e2100499, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35481906

ABSTRACT

Difficulties in obtaining and maintaining the desired level of the critical quality attributes (CQAs) of therapeutic proteins as well as the pace of the development are major challenges of current biopharmaceutical development. Therapeutic proteins, both innovative and biosimilars, are mostly glycosylated. Glycans directly influence the stability, potency, plasma half-life, immunogenicity, and effector functions of the therapeutic. Hence, glycosylation is widely recognized as a process-dependent CQA of therapeutic glycoproteins. Due to the typically high heterogeneity of glycoforms attached to the proteins, control of glycosylation represents one of the most challenging aspects of biopharmaceutical development. Here, we explored a new glycoengineering approach in therapeutic glycoproteins development, which enabled us to achieve the targeted glycoprofile of the Fc-fusion protein in a fast manner. Coupling CRISPRi technology with lectin-FACS sorting enabled downregulation of the endogenous gene involved in fucosylation and further enrichment of CHO cells producing Fc-fusion proteins with reduced fucosylation levels. Enrichment of cells with targeted glycoprofile can lead to time-optimized clone screening and speed up cell line development. Moreover, the presented approach allows isolation of clones with varying levels of fucosylation, which makes it applicable to a broad range of glycoproteins differing in target fucosylation level.


Subject(s)
Biosimilar Pharmaceuticals , Animals , CHO Cells , Clustered Regularly Interspaced Short Palindromic Repeats , Cricetinae , Cricetulus , Glycoproteins/genetics , Polysaccharides
2.
Metab Eng ; 70: 23-30, 2022 03.
Article in English | MEDLINE | ID: mdl-35007751

ABSTRACT

Current protocols for generating stable transgenic cell lines mostly rely on antibiotic selection or the use of specialized cell lines lacking an essential part of their metabolic machinery, but these approaches require working with either toxic chemicals or knockout cell lines, which can reduce productivity. Since most mammalian cells cannot utilize cellobiose, a disaccharide consisting of two ß-1,4-linked glucose molecules, we designed an antibiotic-free selection system, CelloSelect, which consists of a selection cassette encoding Neurospora crassa cellodextrin transporter CDT1 and ß-glucosidase GH1-1. When cultivated in glucose-free culture medium containing cellobiose, CelloSelect-transfected cells proliferate by metabolizing cellobiose as a primary energy source, and are protected from glucose starvation. We show that the combination of CelloSelect with a PiggyBac transposase-based integration strategy provides a platform for the swift and efficient generation of stable transgenic cell lines. Growth rate analysis of metabolically engineered cells in cellobiose medium confirmed the expansion of cells stably expressing high levels of a cargo fluorescent marker protein. We further validated this strategy by applying the CelloSelect system for stable integration of sequences encoding two biopharmaceutical proteins, erythropoietin and the monoclonal antibody rituximab, and confirmed that the proteins are efficiently produced in either cellobiose- or glucose-containing medium in suspension-adapted CHO cells cultured in chemically defined media. We believe coupling heterologous metabolic pathways additively to the endogenous metabolism of mammalian cells has the potential to complement or to replace current cell-line selection systems.


Subject(s)
Cellobiose , Saccharomyces cerevisiae , Animals , Cell Line , Cellobiose/metabolism , Cricetinae , Cricetulus , Fermentation , Metabolic Networks and Pathways , Saccharomyces cerevisiae/metabolism
3.
ACS Synth Biol ; 9(11): 2964-2970, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33213155

ABSTRACT

Synthetic biology relies on rapid and efficient methods to stably integrate DNA payloads encoding for synthetic biological systems into the genome of living cells. The size of designed biological systems increases with their complexity, and novel methods are needed that enable efficient and simultaneous integration of multiple payloads into single cells. By assembling natural and synthetic protein-protein dimerization domains, we have engineered a set of multipartite transcription factors for driving heterologous target gene expression. With the distribution of single parts of multipartite transcription factors on piggyback transposon-based donor plasmids, we have created a logic genome integration control (LOGIC) system that allows for efficient one-step selection of stable mammalian cell lines with up to three plasmids. LOGIC significantly enhances the efficiency of multiplexed payload integration in mammalian cells compared to traditional cotransfection and may advance cell line engineering in synthetic biology and biotechnology.


Subject(s)
Genome/genetics , Mammals/genetics , Transcription Factors/genetics , Animals , Biotechnology/methods , Cell Line , Gene Expression/genetics , Genetic Engineering/methods , Logic , Plasmids/genetics , Protein Domains/genetics , Synthetic Biology/methods
4.
Nat Commun ; 9(1): 1822, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29739943

ABSTRACT

Quorum sensing is a promising target for next-generation anti-infectives designed to address evolving bacterial drug resistance. The autoinducer-2 (AI-2) is a key quorum-sensing signal molecule which regulates bacterial group behaviors and is recognized by many Gram-negative and Gram-positive bacteria. Here we report a synthetic mammalian cell-based microbial-control device that detects microbial chemotactic formyl peptides through a formyl peptide sensor (FPS) and responds by releasing AI-2. The microbial-control device was designed by rewiring an artificial receptor-based signaling cascade to a modular biosynthetic AI-2 production platform. Mammalian cells equipped with the microbial-control gene circuit detect formyl peptides secreted from various microbes with high sensitivity and respond with robust AI-2 production, resulting in control of quorum sensing-related behavior of pathogenic Vibrio harveyi and attenuation of biofilm formation by the human pathogen Candida albicans. The ability to manipulate mixed microbial populations through fine-tuning of AI-2 levels may provide opportunities for future anti-infective strategies.


Subject(s)
Biofilms , Biosensing Techniques , Cell Engineering , Homoserine/analogs & derivatives , Lactones/metabolism , Quorum Sensing/genetics , Vibrio/physiology , Animals , Bacterial Proteins/genetics , Candida albicans/metabolism , Cell Line , Drug Resistance, Microbial , Genes, Bacterial , Homoserine/metabolism , Humans , Receptors, Formyl Peptide/metabolism , Reproducibility of Results , Signal Transduction , Synthetic Biology , Vibrio/genetics , Vibrio/pathogenicity
5.
Nat Methods ; 15(1): 57-60, 2018 01.
Article in English | MEDLINE | ID: mdl-29200199

ABSTRACT

Synthetic biologists have advanced the design of trigger-inducible gene switches and their assembly into input-programmable circuits that enable engineered human cells to perform arithmetic calculations reminiscent of electronic circuits. By designing a versatile plug-and-play molecular-computation platform, we have engineered nine different cell populations with genetic programs, each of which encodes a defined computational instruction. When assembled into 3D cultures, these engineered cell consortia execute programmable multicellular full-adder logics in response to three trigger compounds.


Subject(s)
Cell Culture Techniques/methods , Gene Regulatory Networks , Software , Synthetic Biology/methods , Genes, Reporter , Humans
6.
Nat Commun ; 8(1): 1101, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29062109

ABSTRACT

While constantly rising, the prevalence of allergies is globally one of the highest among chronic diseases. Current treatments of allergic diseases include the application of anti-histamines, immunotherapy, steroids, and anti-immunoglobulin E (IgE) antibodies. Here we report mammalian cells engineered with a synthetic signaling cascade able to monitor extracellular pathophysiological levels of interleukin 4 and interleukin 13, two main cytokines orchestrating allergic inflammation. Upon activation of transgenic cells by these cytokines, designed ankyrin repeat protein (DARPin) E2_79, a non-immunogenic protein binding human IgE, is secreted in a precisely controlled and reversible manner. Using human whole blood cell culturing, we demonstrate that the mammalian dual T helper 2 cytokine sensor produces sufficient levels of DARPin E2_79 to dampen histamine release in allergic subjects exposed to allergens. Hence, therapeutic gene networks monitoring disease-associated cytokines coupled with in situ production, secretion and systemic delivery of immunomodulatory biologics may foster advances in the treatment of allergies.


Subject(s)
Hypersensitivity/genetics , Interleukin-13/immunology , Interleukin-4/immunology , Recombinant Fusion Proteins/genetics , Allergens/immunology , Cell Line , Genetic Engineering , Histamine/immunology , Histamine/metabolism , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Interleukin-13/genetics , Interleukin-4/genetics , Protein Binding , Recombinant Fusion Proteins/metabolism , Th2 Cells/immunology
7.
Nat Chem Biol ; 13(3): 309-316, 2017 03.
Article in English | MEDLINE | ID: mdl-28092361

ABSTRACT

Synthetic biology advances the rational engineering of mammalian cells to achieve cell-based therapy goals. Synthetic gene networks have nearly reached the complexity of digital electronic circuits and enable single cells to perform programmable arithmetic calculations or to provide dynamic remote control of transgenes through electromagnetic waves. We designed a synthetic multilayered gaseous-fragrance-programmable analog-to-digital converter (ADC) allowing for remote control of digital gene expression with 2-bit AND-, OR- and NOR-gate logic in synchronized cell consortia. The ADC consists of multiple sampling-and-quantization modules sensing analog gaseous fragrance inputs; a gas-to-liquid transducer converting fragrance intensity into diffusible cell-to-cell signaling compounds; a digitization unit with a genetic amplifier circuit to improve the signal-to-noise ratio; and recombinase-based digital expression switches enabling 2-bit processing of logic gates. Synthetic ADCs that can remotely control cellular activities with digital precision may enable the development of novel biosensors and may provide bioelectronic interfaces synchronizing analog metabolic pathways with digital electronics.


Subject(s)
Analog-Digital Conversion , Odorants/analysis , Synthetic Biology/methods , Cells, Cultured , HEK293 Cells , Humans , Perfume/analysis
8.
Angew Chem Int Ed Engl ; 56(23): 6396-6419, 2017 06 01.
Article in English | MEDLINE | ID: mdl-27943572

ABSTRACT

Synthetic biology concerns the engineering of man-made living biomachines from standardized components that can perform predefined functions in a (self-)controlled manner. Different research strategies and interdisciplinary efforts are pursued to implement engineering principles to biology. The "top-down" strategy exploits nature's incredible diversity of existing, natural parts to construct synthetic compositions of genetic, metabolic, or signaling networks with predictable and controllable properties. This mainly application-driven approach results in living factories that produce drugs, biofuels, biomaterials, and fine chemicals, and results in living pills that are based on engineered cells with the capacity to autonomously detect and treat disease states in vivo. In contrast, the "bottom-up" strategy seeks to be independent of existing living systems by designing biological systems from scratch and synthesizing artificial biological entities not found in nature. This more knowledge-driven approach investigates the reconstruction of minimal biological systems that are capable of performing basic biological phenomena, such as self-organization, self-replication, and self-sustainability. Moreover, the syntheses of artificial biological units, such as synthetic nucleotides or amino acids, and their implementation into polymers inside living cells currently set the boundaries between natural and artificial biological systems. In particular, the in vitro design, synthesis, and transfer of complete genomes into host cells point to the future of synthetic biology: the creation of designer cells with tailored desirable properties for biomedicine and biotechnology.


Subject(s)
Synthetic Biology , Animals , Biocompatible Materials , Biofuels , Cell Engineering , Cell- and Tissue-Based Therapy , Cell-Free System , Genetic Engineering , Genome , Humans , Interdisciplinary Studies
9.
Nucleic Acids Res ; 44(10): e94, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26939886

ABSTRACT

Hammerhead ribozymes are self-cleaving RNA molecules capable of regulating gene expression in living cells. Their cleavage performance is strongly influenced by intra-molecular loop-loop interactions, a feature not readily accessible through modern prediction algorithms. Ribozyme engineering and efficient implementation of ribozyme-based genetic switches requires detailed knowledge of individual self-cleavage performances. By rational design, we devised fluorescent aptamer-ribozyme RNA architectures that allow for the real-time measurement of ribozyme self-cleavage activity in vitro The engineered nucleic acid molecules implement a split Spinach aptamer sequence that is made accessible for strand displacement upon ribozyme self-cleavage, thereby complementing the fluorescent Spinach aptamer. This fully RNA-based ribozyme performance assay correlates ribozyme cleavage activity with Spinach fluorescence to provide a rapid and straightforward technology for the validation of loop-loop interactions in hammerhead ribozymes.


Subject(s)
Aptamers, Nucleotide/chemistry , Genetic Complementation Test/methods , RNA, Catalytic/metabolism , Fluorescent Dyes/chemistry , Genetic Engineering/methods , RNA, Catalytic/genetics
10.
Mol Cell Biol ; 36(1): 132-43, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26483415

ABSTRACT

Death-associated protein kinase 2 (DAPK2) is a Ca(2+)/calmodulin-dependent Ser/Thr kinase that possesses tumor-suppressive functions and regulates programmed cell death, autophagy, oxidative stress, hematopoiesis, and motility. As only few binding partners of DAPK2 have been determined, the molecular mechanisms governing these biological functions are largely unknown. We report the identification of 180 potential DAPK2 interaction partners by affinity purification-coupled mass spectrometry, 12 of which are known DAPK binding proteins. A small subset of established and potential binding proteins detected in this screen was further investigated by bimolecular fluorescence complementation (BiFC) assays, a method to visualize protein interactions in living cells. These experiments revealed that α-actinin-1 and 14-3-3-ß are novel DAPK2 binding partners. The interaction of DAPK2 with α-actinin-1 was localized at the plasma membrane, resulting in massive membrane blebbing and reduced cellular motility, whereas the interaction of DAPK2 with 14-3-3-ß was localized to the cytoplasm, with no impact on blebbing, motility, or viability. Our results therefore suggest that DAPK2 effector functions are influenced by the protein's subcellular localization and highlight the utility of combining mass spectrometry screening with bimolecular fluorescence complementation to identify and characterize novel protein-protein interactions.


Subject(s)
Apoptosis/physiology , Death-Associated Protein Kinases/metabolism , Fluorescence , Proteomics , Autophagy/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Humans , Protein Binding/physiology , Proteomics/methods
11.
Nat Methods ; 11(11): 1154-60, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25282610

ABSTRACT

RNAs are ideal for the design of gene switches that can monitor and program cellular behavior because of their high modularity and predictable structure-function relationship. We have assembled an expression platform with an embedded modular ribozyme scaffold that correlates self-cleavage activity of designer ribozymes with transgene translation in bacteria and mammalian cells. A design approach devised to screen ribozyme libraries in bacteria and validate variants with functional tertiary stem-loop structures in mammalian cells resulted in a designer ribozyme with a protein-binding nutR-boxB stem II and a selected matching stem I. In a mammalian expression context, this designer ribozyme exhibited dose-dependent translation control by the N-peptide, had rapid induction kinetics and could be combined with classic small molecule-responsive transcription control modalities to construct complex, programmable genetic circuits.


Subject(s)
Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/genetics , Gene Regulatory Networks , Isoenzymes/biosynthesis , Isoenzymes/genetics , Protein Biosynthesis , RNA, Catalytic/metabolism , Riboswitch , Transgenes , Animals , Binding Sites/genetics , CHO Cells , Cricetulus , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Gene Expression , Green Fluorescent Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Catalytic/chemistry , RNA, Catalytic/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Structure-Activity Relationship , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Nat Commun ; 5: 4408, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25093291

ABSTRACT

Allergic disorders are markedly increasing in industrialized countries. The identification of compounds that trigger the immunoglobulin E-dependent allergic reaction remains the key to limit patients' exposure to critical allergens and improve their quality of life. Here we use synthetic biology principles to design a mammalian cell-based allergy profiler that scores the allergen-triggered release of histamine from whole-blood-derived human basophils. A synthetic signalling cascade engineered within the allergy profiler rewires histamine input to the production of reporter protein, thereby integrating histamine levels in whole-blood samples with remarkable sensitivity and a wide dynamic range, allowing for rapid results or long-term storage of output, respectively. This approach provides non-intrusive allergy profiles for the personalized medicine era.


Subject(s)
Histamine Release/immunology , Histamine/chemistry , Hypersensitivity/immunology , Precision Medicine/methods , Synthetic Biology , Allergens/chemistry , Animals , Basophils/cytology , Basophils/metabolism , CHO Cells , Cricetulus , Gene Expression Regulation , Genes, Reporter , HEK293 Cells , Humans , Immunoglobulin E/immunology , Pollen/chemistry , Quality of Life , Transgenes
13.
Mol Cell ; 55(3): 397-408, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25018017

ABSTRACT

All metabolic activities operate within a narrow pH range that is controlled by the CO2-bicarbonate buffering system. We hypothesized that pH could serve as surrogate signal to monitor and respond to the physiological state. By functionally rewiring the human proton-activated cell-surface receptor TDAG8 to chimeric promoters, we created a synthetic signaling cascade that precisely monitors extracellular pH within the physiological range. The synthetic pH sensor could be adjusted by organic acids as well as gaseous CO2 that shifts the CO2-bicarbonate balance toward hydrogen ions. This enabled the design of gas-programmable logic gates, provided remote control of cellular behavior inside microfluidic devices, and allowed for CO2-triggered production of biopharmaceuticals in standard bioreactors. When implanting cells containing the synthetic pH sensor linked to production of insulin into type 1 diabetic mice developing diabetic ketoacidosis, the prosthetic network automatically scored acidic pH and coordinated an insulin expression response that corrected ketoacidosis.


Subject(s)
Carbon Dioxide/metabolism , Diabetic Ketoacidosis/physiopathology , Microfluidic Analytical Techniques/methods , Receptors, G-Protein-Coupled/genetics , Synthetic Biology/methods , Animals , CHO Cells , Cell Line , Cell Transplantation , Cricetulus , Diabetic Ketoacidosis/therapy , Disease Models, Animal , Female , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Mice , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 111(22): 8155-60, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24830426

ABSTRACT

Sharks and other cartilaginous fish are the phylogenetically oldest living organisms that rely on antibodies as part of their adaptive immune system. They produce the immunoglobulin new antigen receptor (IgNAR), a homodimeric heavy chain-only antibody, as a major part of their humoral adaptive immune response. Here, we report the atomic resolution structure of the IgNAR constant domains and a structural model of this heavy chain-only antibody. We find that despite low sequence conservation, the basic Ig fold of modern antibodies is already present in the evolutionary ancient shark IgNAR domains, highlighting key structural determinants of the ubiquitous Ig fold. In contrast, structural differences between human and shark antibody domains explain the high stability of several IgNAR domains and allowed us to engineer human antibodies for increased stability and secretion efficiency. We identified two constant domains, C1 and C3, that act as dimerization modules within IgNAR. Together with the individual domain structures and small-angle X-ray scattering, this allowed us to develop a structural model of the complete IgNAR molecule. Its constant region exhibits an elongated shape with flexibility and a characteristic kink in the middle. Despite the lack of a canonical hinge region, the variable domains are spaced appropriately wide for binding to multiple antigens. Thus, the shark IgNAR domains already display the well-known Ig fold, but apart from that, this heavy chain-only antibody employs unique ways for dimerization and positioning of functional modules.


Subject(s)
Antibodies/blood , Evolution, Molecular , Osmoregulation/immunology , Receptors, Antigen/metabolism , Sharks/immunology , Adaptive Immunity/physiology , Amino Acid Sequence , Animals , Antibodies/chemistry , Cells, Cultured , Humans , Immunoglobulin Constant Regions/chemistry , Immunoglobulin Constant Regions/genetics , Immunoglobulin Constant Regions/metabolism , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Insecta , Molecular Sequence Data , Protein Engineering , Protein Folding , Protein Stability , Protein Structure, Tertiary , Receptors, Antigen/chemistry , Receptors, Antigen/genetics , Sharks/physiology , Urea/metabolism
15.
Apoptosis ; 18(9): 1093-105, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23733106

ABSTRACT

Noxa is a member of the pro-apoptotic BH3-only group of Bcl-2 proteins that is known to bind specifically to anti-apoptotic Mcl-1 and A1, antagonizing their function. Mcl-1 has been reported to have a short half-life, and Noxa up-regulation accelerates Mcl-1 degradation by the proteasome. Unlike human Noxa, mouse Noxa has two BH3-domains, which both have affinity for Mcl-1. We here investigate two aspects of the molecular function of Noxa, namely the requirements for the two BH3-domains in mouse Noxa and the role of Noxa in Mcl-1-degradation. We found that only the C-terminal BH3-domain of mouse Noxa is active in neutralizing Mcl-1. This was the result of the targeting of Noxa to the outer mitochondrial membrane through its C-terminal alpha-helix, which allowed Mcl-1-neutralization only when the BH3-domain was immediately N-terminal of the membrane anchor. However, the N-terminal BH3-domain enhanced interaction with Mcl-1 and A1. The Noxa-dependent degradation of Mcl-1 was independent of the kinase GSK3 and the deubiquitinase Usp9x in mouse embryonic fibroblasts. These data show that Noxa is targeted to the mitochondrial membrane where it neutralises Mcl-1 via its C-terminal BH3-domain and suggest that Noxa is co-degraded with Noxa, in a way independent of ubiquitin-modifying enzymes described for Mcl-1.


Subject(s)
Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Humans , Mice , Mitochondrial Membranes/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Protein Binding , Protein Structure, Tertiary , Protein Transport , Proteolysis , Proto-Oncogene Proteins c-bcl-2/genetics
17.
Nature ; 487(7405): 123-7, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22722847

ABSTRACT

Synthetic biology has advanced the design of standardized control devices that program cellular functions and metabolic activities in living organisms. Rational interconnection of these synthetic switches resulted in increasingly complex designer networks that execute input-triggered genetic instructions with precision, robustness and computational logic reminiscent of electronic circuits. Using trigger-controlled transcription factors, which independently control gene expression, and RNA-binding proteins that inhibit the translation of transcripts harbouring specific RNA target motifs, we have designed a set of synthetic transcription­translation control devices that could be rewired in a plug-and-play manner. Here we show that these combinatorial circuits integrated a two-molecule input and performed digital computations with NOT, AND, NAND and N-IMPLY expression logic in single mammalian cells. Functional interconnection of two N-IMPLY variants resulted in bitwise intracellular XOR operations, and a combinatorial arrangement of three logic gates enabled independent cells to perform programmable half-subtractor and half-adder calculations. Individual mammalian cells capable of executing basic molecular arithmetic functions isolated or coordinated to metabolic activities in a predictable, precise and robust manner may provide new treatment strategies and bio-electronic interfaces in future gene-based and cell-based therapies.


Subject(s)
Computers, Molecular , Gene Expression Regulation , Single-Cell Analysis , Synthetic Biology/methods , Cell- and Tissue-Based Therapy , Gene Regulatory Networks , Genetic Therapy , HEK293 Cells , Humans , Logic , Mathematics , Transcription Factors/metabolism
18.
Metab Eng ; 14(4): 325-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22543310

ABSTRACT

The biosynthesis of non-ribosomal peptides, many of which have pharmaceutical activities, is an evolutionary privilege of microorganisms. Capitalizing on the universal set of the Streptomyces lavendulae non-ribosomal peptide synthase BpsA and the Streptomyces verticillus 4'-phosphopantetheinyl transferase Svp, we have engineered Escherichia coli as well as mammalian cells, including human stem cells, to produce the blue 3,3'-bipyridyl pigment keto-indigoidine and the reduced colorless but fluorescent leuco-isoform. Detailed characterization of a tailored substrate-free chromogenic assay and FACS analysis showed that indigoidine's blue color and fluorescence could be reliably profiled in bacteria and mammalian cells using standard multiwell-compatible detection equipment. Besides serving as an inexpensive, reliable, versatile and easy-to-assay cross-kingdom reporter system, the potential of having mammalian cells produce non-ribosomal peptides, preferably ones with biopharmaceutical activities, may provide novel treatment opportunities in future gene- and cell-based therapies.


Subject(s)
Piperidones/metabolism , Streptomyces/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Embryonic Stem Cells , Escherichia coli/metabolism , Genes, Reporter , Humans , Metabolic Engineering/methods , Peptide Synthases/genetics , Peptide Synthases/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism
19.
Methods ; 56(3): 351-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22305857

ABSTRACT

Artificial RNA riboswitches--apart from protein-based gene regulation systems, which have been known about for a long time--have become increasingly important in biotechnology and synthetic biology. Aptamer-controlled hammerhead ribozymes (so-called aptazymes) have been shown to be a versatile platform for the engineering of novel gene regulators. Since aptazymes are cis-acting elements that are located in the untranslated regions of a gene of interest, their application does not need any further protein co-factor. This presents the opportunity to simplify complex gene networks while simultaneously expanding the repertoire of available parts. Nevertheless, the generation of novel aptazymes requires a functional aptamer-ribozyme connection, which can be difficult to engineer. This article describes a novel approach for using fluorescence activated cell sorting (FACS) in order to identify functional aptazymes in bacteria and their subsequent transfer into mammalian cells.


Subject(s)
Genetic Engineering , RNA, Catalytic/physiology , Riboswitch , Amino Acid Sequence , Animals , Aptamers, Nucleotide/genetics , Humans , Molecular Sequence Data
20.
Nucleic Acids Res ; 39(22): e155, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21984476

ABSTRACT

Aptamers binding proteins or small molecules have been shown to be versatile and powerful building blocks for the construction of artificial genetic switches. In this study, we present a novel aptamer-based construct regulating the Tet Off system in a tetracycline-independent manner thus achieving control of transgene expression. For this purpose, a TetR protein-inhibiting aptamer was engineered for use in mammalian cells, enabling the RNA-responsive control of the tetracycline-dependent transactivator (tTA). By rationally attaching the theophylline aptamer as a sensor, the inhibitory TetR aptamer and thus tTA activity became dependent on the ligand of the sensor aptamer. Addition of the small molecule theophylline resulted in enhanced binding to the corresponding protein in vitro and in inhibition of reporter gene expression in mammalian cell lines. By using aptamers as adaptors in order to control protein activity by a predetermined small molecule, we present a simple and straightforward approach for future applications in the field of Chemical Biology. Moreover, aptamer-based control of the widely used Tet system introduces a new layer of regulation thereby facilitating the construction of more complex gene networks.


Subject(s)
Aptamers, Nucleotide/chemistry , Gene Expression Regulation , Transgenes , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Molecular Sequence Data , Repressor Proteins/metabolism , Theophylline/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...