19 results on '"Leo Scheller"'
Search Results
2. Phosphoregulated orthogonal signal transduction in mammalian cells
- Author
-
Leo Scheller, Marc Schmollack, Adrian Bertschi, Maysam Mansouri, Pratik Saxena, and Martin Fussenegger
- Subjects
Science - Abstract
Phosphoregulation is a key mechanism of signal processing. Here the authors build a phosphoregulated relay system in mammalian cells for orthogonal signal transduction.
- Published
- 2020
- Full Text
- View/download PDF
3. Author Correction: A modular degron library for synthetic circuits in mammalian cells
- Author
-
Hélène Chassin, Marius Müller, Marcel Tigges, Leo Scheller, Moritz Lang, and Martin Fussenegger
- Subjects
Science - Published
- 2023
- Full Text
- View/download PDF
4. Rewiring of endogenous signaling pathways to genomic targets for therapeutic cell reprogramming
- Author
-
Krzysztof Krawczyk, Leo Scheller, Hyojin Kim, and Martin Fussenegger
- Subjects
Science - Abstract
Current cellular rewiring designs are typically tailored to detect single inputs. Here the authors present GEARs that function independently of engineered receptor/reporter systems and directly reroute endogenous signaling pathways to alternative genomic loci using dCas9-directed gene expression.
- Published
- 2020
- Full Text
- View/download PDF
5. A modular degron library for synthetic circuits in mammalian cells
- Author
-
Hélène Chassin, Marius Müller, Marcel Tigges, Leo Scheller, Moritz Lang, and Martin Fussenegger
- Subjects
Science - Abstract
One method of controlling protein degradation is through the use of degrons. Here the authors present a toolbox of characterised degrons as a library to fine-tune biological gene-expression systems. Its application is demonstrated by a set of tunable synthetic pulse generators in mammalian cells.
- Published
- 2019
- Full Text
- View/download PDF
6. Caffeine-inducible gene switches controlling experimental diabetes
- Author
-
Daniel Bojar, Leo Scheller, Ghislaine Charpin-El Hamri, Mingqi Xie, and Martin Fussenegger
- Subjects
Science - Abstract
Control of transgene expression should ideally be easy and with minimal side effects. Here the authors present a synthetic biology-based approach in which the caffeine in coffee regulates a genetic circuit controlling glucagon-like peptide 1 expression in diabetic mice.
- Published
- 2018
- Full Text
- View/download PDF
7. Rationally designed protein bandpass filters for controlling cellular signaling with chemical inputs
- Author
-
Sailan Shui, Leo Scheller, and Bruno E. Correia
- Abstract
Biological mechanisms that rely on signal integration and processing are fundamental for cell function. These types of capabilities are analogous to those found in electronic circuits where individual components perform operations on input signals. In electronics, bandpass filters are crucial components to narrow frequencies within a specified range and reject frequencies outside of that range. However, no generalizable protein-based components are currently available to mimic such processes in engineered biological systems, representing an unmet need in controllable modules. Here, we propose a rational design approach to create protein-basedchemically responsive bandpass filters(CBP) which pass chemical concentrations within a range and reject concentrations outside of that range, showing an OFF-ON-OFF regulatory pattern. The CBPs were designed using structure-based approaches where we created a heterodimeric construct which the assembly is triggered by low concentration of a small-molecule, and this interaction is inhibited at high concentrations of the drug, effectively creating a bandpass filter. The CBPs have a multidomain architecture where we used known drug-receptors, a computationally designed protein binder and small-molecule inhibitors. Owing to the modularity of the system, each domain of the CBPs can be rationally fine-tuned to optimize its performance, including bandwidth, maximum response, cutoff concentration and fold changes. These CBPs were used to regulate cell surface receptor signaling pathways showing the capability to control cellular activities in engineered cells.
- Published
- 2023
- Full Text
- View/download PDF
8. Rational design of chemically controlled antibodies and protein therapeutics
- Author
-
Anthony Marchand, Lucia Bonati, Sailan Shui, Leo Scheller, Pablo Gainza, Stéphane Rosset, Sandrine Georgeon, Li Tang, and Bruno E. Correia
- Subjects
computational ,antibody ,therapeutics ,small molecule ,antibodies ,drug ,protein ,protein design ,switch - Abstract
Protein-based therapeutics such as monoclonal antibodies and cytokines are important therapies in various pathophysiological conditions such as oncology, auto-immune disorders, and viral infections. However, the wide application of such protein therapeutics is often hindered by dose-limiting toxicities and adverse effects, namely cytokine storm syndrome, organ failure and others. Therefore, spatiotemporal control of the activities of these proteins is crucial to further expand their application. Here, we report the design and application of small molecule-controlled switchable protein therapeutics by taking advantage of a previously engineered OFF-switch system. We used Rosetta modeling suite to computationally optimize the affinity between B-cell lymphoma 2 (Bcl-2) protein and a previously developed computationally designed protein partner (LD3) to obtain a fast and efficient heterodimer disruption upon addition of a competing drug (Venetoclax). The incorporation of the engineered OFF-switch system intoαCTLA4, anti-HER2 antibodies or an Fc-fused IL-15 cytokine demonstrated an efficient disruption in vitro, as well as fast clearance in vivo upon addition of the competing drug Venetoclax. These results provide a proof-of-concept for the rational design of controllable biologics by introducing a drug-induced OFF-switch into existing protein-based therapeutics.
- Published
- 2022
- Full Text
- View/download PDF
9. Bottom-up de novo design of functional proteins with complex structural features
- Author
-
Sandrine Georgeon, Leo Scheller, Fabian Sesterhenn, Bruno E. Correia, Stéphane Rosset, Jaume Bonet, Martin Fussenegger, Theodore S. Jardetzky, Thomas Krey, Maarten Merkx, Johannes T. Cramer, Luciano A. Abriata, Xiaolin Wen, Che Yang, Eva A. van Aalen, Group Den Toonder, Protein Engineering, and ICMS Core
- Subjects
0303 health sciences ,Chemistry ,motif ,030302 biochemistry & molecular biology ,Protein design ,enzymes ,computational design ,fold ,rsv ,tool ,Cell Biology ,Computational biology ,fusion glycoprotein ,principles ,03 medical and health sciences ,Protein structure ,Molecular recognition ,Antibody response ,proof ,Computational design ,antibodies ,Protein folding ,Structural motif ,Molecular Biology ,030304 developmental biology - Abstract
De novo protein design has enabled the creation of new protein structures. However, the design of functional proteins has proved challenging, in part due to the difficulty of transplanting structurally complex functional sites to available protein structures. Here, we used a bottom-up approach to build de novo proteins tailored to accommodate structurally complex functional motifs. We applied the bottom-up strategy to successfully design five folds for four distinct binding motifs, including a bifunctionalized protein with two motifs. Crystal structures confirmed the atomic-level accuracy of the computational designs. These de novo proteins were functional as components of biosensors to monitor antibody responses and as orthogonal ligands to modulate synthetic signaling receptors in engineered mammalian cells. Our work demonstrates the potential of bottom-up approaches to accommodate complex structural motifs, which will be essential to endow de novo proteins with elaborate biochemical functions, such as molecular recognition or catalysis. [Figure not available: see fulltext.]
- Published
- 2021
10. Programmable DARPin-based receptors for the detection of thrombotic markers
- Author
-
Tobias Strittmatter, Yidan Wang, Adrian Bertschi, Leo Scheller, Patrick C. Freitag, Preetam Guha Ray, Pascal Stuecheli, Jonas V. Schaefer, Thomas Reinberg, Dimitrios Tsakiris, Andreas Plückthun, Haifeng Ye, Martin Fussenegger, University of Zurich, and Fussenegger, Martin
- Subjects
therapy ,crystal-structure ,selection ,610 Medicine & health ,Cell Biology ,proteins ,Ankyrin Repeat ,1307 Cell Biology ,Fibrin Fibrinogen Degradation Products ,10019 Department of Biochemistry ,1312 Molecular Biology ,570 Life sciences ,biology ,Humans ,Designed Ankyrin Repeat Proteins ,degradation-products ,fibrin ,Carrier Proteins ,Molecular Biology ,binders ,Protein Binding ,Single-Chain Antibodies - Abstract
Cellular therapies remain constrained by the limited availability of sensors for disease markers. Here we present an integrated target-to-receptor pipeline for constructing a customizable advanced modular bispecific extracellular receptor (AMBER) that combines our generalized extracellular molecule sensor (GEMS) system with a high-throughput platform for generating designed ankyrin repeat proteins (DARPins). For proof of concept, we chose human fibrin degradation products (FDPs) as markers with high clinical relevance and screened a DARPin library for FDP binders. We built AMBERs equipped with 19 different DARPins selected from 160 hits, and found 4 of them to be functional as heterodimers with a known single-chain variable fragments binder. Tandem receptors consisting of combinations of the validated DARPins are also functional. We demonstrate applications of these AMBER receptors in vitro and in vivo by constructing designer cell lines that detect pathological concentrations of FDPs and respond with the production of a reporter and a therapeutic anti-thrombotic protein., Nature Chemical Biology, 18, ISSN:1552-4450, ISSN:1552-4469
- Published
- 2021
11. A rational blueprint for the design of chemically-controlled protein switches
- Author
-
Yoichi Kurumida, Sandrine Georgeon, Pablo Gainza, Stéphane Rosset, Leo Scheller, Che Yang, Bruno E. Correia, and Sailan Shui
- Subjects
Drug ,Computer science ,Blueprint ,media_common.quotation_subject ,Immunogenicity ,Protein design ,Computational biology ,Small molecule ,Chemical space ,media_common - Abstract
Small-molecule responsive protein switches are crucial components to control synthetic cellular activities. However, the repertoire of small-molecule protein switches is insufficient for many applications, including those in the translational spaces, where properties such as safety, immunogenicity, drug half-life, and drug side-effects are critical. Here, we present a computational protein design strategy to repurpose drug-inhibited protein-protein interactions as OFF- and ON-switches. The designed binders and drug-receptors form chemically-disruptable heterodimers (CDH) which dissociate in the presence of small molecules. To design ON-switches, we converted the CDHs into a multi-domain architecture which we refer to as activation by inhibitor release switches (AIR) that incorporate a rationally designed drug-insensitive receptor protein. CDHs and AIRs showed excellent performance as drug responsive switches to control combinations of synthetic circuits in mammalian cells. This approach effectively expands the chemical space and logic responses in living cells and provides a blueprint to develop new ON- and OFF-switches for basic and translational applications.
- Published
- 2021
- Full Text
- View/download PDF
12. Synthetic Receptors for Sensing Soluble Molecules with Mammalian Cells
- Author
-
Leo Scheller
- Subjects
Synthetic biology ,Chemistry ,Cell surface receptor ,HEK 293 cells ,Intracellular receptor ,NFAT ,Signal transduction ,Receptor ,Transcription factor ,Cell biology - Abstract
Synthetic receptors control cell behavior in response to environmental stimuli for applications in basic research and cell therapy. However, the integration of synthetic receptors in unexplored contexts is cumbersome, especially for nonspecialist laboratories. Here, I provide a detailed protocol on how to use receptors of the generalized extracellular molecule sensor (GEMS) platform. GEMS is a modular receptor system that can be adapted to sense molecules of choice by using affinity domains that dimerize in response to the target. GEMS consist of an erythropoietin receptor scaffold that has been mutated to no longer bind to erythropoietin. N-terminal fusions with affinity domains, such as single chain variable fragments (scFvs), that bind to two epitopes on the same target activate the receptor. The intracellular receptor domain can be chosen from several signal transduction domains of single-pass transmembrane receptors to activate endogenous signaling pathways. As of now, GEMS have been used for sensing prostate specific antigen (PSA), the synthetic azo dye RR120, caffeine, nicotine, rapamycin, the SunTag peptide, and a de novo designed protein displaying two viral epitopes. The tested intracellular domains were derived from FGFR1, IL-6RB, and VEGFR2, and were used to drive transgene expression from reporter plasmids responsive to the endogenous transcription factors STAT3, NFAT, NF-κB, and a synthetic transcription factor activated by the MAPK pathway. In this protocol, I focus on transient transfections of HEK293T cells and include several general notes about cell handling. While the described methods are optimized for experiments with GEMS, most of the described techniques are general procedures to set up synthetic biology experiments in mammalian cell culture. I outline how to generate stable cell lines and share tips on how to adapt GEMS for new ligands. The main objective of this protocol is to make the GEMS technology accessible also to nonspecialist laboratories to facilitate the use of synthetic receptors in new research contexts.
- Published
- 2021
- Full Text
- View/download PDF
13. A bottom-up approach for the de novo design of functional proteins
- Author
-
Maarten Merkx, Fabian Sesterhenn, Jaume Bonet, Sandrine Georgeon, Leo Scheller, Che Yang, Martin Fussenegger, Theodore S. Jardetzky, Stéphane Rosset, Johannes T. Cramer, Eva A. van Aalen, Luciano A. Abriata, Bruno E. Correia, Xiaolin Wen, and Thomas Krey
- Subjects
0303 health sciences ,Novel protein ,Functional protein ,Chemistry ,Protein design ,Computational biology ,Transplantation ,03 medical and health sciences ,0302 clinical medicine ,Template ,Antibody response ,Signal transduction ,Protein secondary structure ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
De novo protein design has enabled the creation of novel protein structures. To design novel functional proteins, state-of-the-art approaches use natural proteins or first design protein scaffolds that subsequently serve as templates for the transplantation of functional motifs. In these approaches, the templates are function-agnostic and motifs have been limited to those with regular secondary structure. Here, we present a bottom-up approach to build de novo proteins tailored to structurally complex functional motifs. We applied a bottom-up strategy to design scaffolds for four different binding motifs, including one bi-functionalized protein with two motifs. The de novo proteins were functional as biosensors to quantify epitope-specific antibody responses and as orthogonal ligands to activate a signaling pathway in engineered mammalian cells. Altogether, we present a versatile strategy for the bottom-up design of functional proteins, applicable to a wide range of functional protein design challenges.
- Published
- 2020
- Full Text
- View/download PDF
14. Generalized extracellular molecule sensor platform for programming cellular behavior
- Author
-
David W Fuchs, Leo Scheller, Tobias Strittmatter, Daniel Bojar, and Martin Fussenegger
- Subjects
Male ,0301 basic medicine ,MAPK/ERK pathway ,Nicotine ,030102 biochemistry & molecular biology ,Chemistry ,Transgene ,HEK 293 cells ,Prostatic Neoplasms ,Cell Biology ,Prostate-Specific Antigen ,Small molecule ,stat ,Cell biology ,03 medical and health sciences ,HEK293 Cells ,030104 developmental biology ,Biomarkers, Tumor ,Extracellular ,Humans ,Receptor ,Molecular Biology ,Cells, Cultured ,PI3K/AKT/mTOR pathway ,Fluorescent Dyes - Abstract
Strategies for expanding the sensor space of designer receptors are urgently needed to tailor cell-based therapies to respond to any type of medically relevant molecules. Here, we describe a universal approach to designing receptor scaffolds that enables antibody-specific molecular input to activate JAK/STAT, MAPK, PLCG or PI3K/Akt signaling rewired to transgene expression driven by synthetic promoters. To demonstrate its scope, we equipped the GEMS (generalized extracellular molecule sensor) platform with antibody fragments targeting a synthetic azo dye, nicotine, a peptide tag and the PSA (prostate-specific antigen) biomarker, thereby covering inputs ranging from small molecules to proteins. These four GEMS devices provided robust signaling and transgene expression with high signal-to-noise ratios in response to their specific ligands. The sensitivity of the nicotine- and PSA-specific GEMS devices matched the clinically relevant concentration ranges, and PSA-specific GEMS were able to detect pathological PSA levels in the serum of patients diagnosed with prostate cancer.
- Published
- 2018
- Full Text
- View/download PDF
15. A modular degron library for synthetic circuits in mammalian cells
- Author
-
Martin Fussenegger, Marcel Tigges, Leo Scheller, Moritz Lang, Marius Müller, and Hélène Chassin
- Subjects
0301 basic medicine ,Intravital Microscopy ,Computer science ,Science ,General Physics and Astronomy ,02 engineering and technology ,Computational biology ,Protein degradation ,Protein Engineering ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Humans ,Amino Acid Sequence ,lcsh:Science ,Gene ,Electronic circuit ,Regulation of gene expression ,Multidisciplinary ,business.industry ,HEK 293 cells ,Mesenchymal Stem Cells ,General Chemistry ,Modular design ,021001 nanoscience & nanotechnology ,030104 developmental biology ,HEK293 Cells ,Gene Expression Regulation ,Microscopy, Fluorescence ,Proteolysis ,lcsh:Q ,Synthetic Biology ,Target protein ,Degron ,0210 nano-technology ,business ,Biotechnology ,Half-Life ,HeLa Cells - Abstract
Tight control over protein degradation is a fundamental requirement for cells to respond rapidly to various stimuli and adapt to a fluctuating environment. Here we develop a versatile, easy-to-handle library of destabilizing tags (degrons) for the precise regulation of protein expression profiles in mammalian cells by modulating target protein half-lives in a predictable manner. Using the well-established tetracycline gene-regulation system as a model, we show that the dynamics of protein expression can be tuned by fusing appropriate degron tags to gene regulators. Next, we apply this degron library to tune a synthetic pulse-generating circuit in mammalian cells. With this toolbox we establish a set of pulse generators with tailored pulse lengths and magnitudes of protein expression. This methodology will prove useful in the functional roles of essential proteins, fine-tuning of gene-expression systems, and enabling a higher complexity in the design of synthetic biological systems in mammalian cells., Nature Communications, 10 (1), ISSN:2041-1723
- Published
- 2019
16. Genetically encoded betaxanthin-based small-molecular fluorescent reporter for mammalian cells
- Author
-
Martin Fussenegger, David W Fuchs, Tobias Strittmatter, Pascal Stücheli, Pratik Saxena, Simon Sieber, Leo Scheller, Karl Gademann, University of Zurich, and Fussenegger, Martin
- Subjects
10120 Department of Chemistry ,AcademicSubjects/SCI00010 ,dopa-dioxygenase ,01 natural sciences ,stress ,Narese/2 ,Genes, Reporter ,Narese/1 ,Cricetinae ,540 Chemistry ,Gene expression ,Picolinic Acids ,time ,chemistry.chemical_classification ,0303 health sciences ,Chinese hamster ovary cell ,Absorption, Radiation ,Recombinant Proteins ,Biochemistry ,Oxygenases ,Methods Online ,Single-Cell Analysis ,Tyrosine 3-Monooxygenase ,Ultraviolet Rays ,proliferation ,pigments ,Heterologous ,CHO Cells ,Biology ,Fungal Proteins ,03 medical and health sciences ,Cricetulus ,1311 Genetics ,Genetics ,Animals ,Humans ,Viability assay ,030304 developmental biology ,010405 organic chemistry ,HEK 293 cells ,gene-expression ,proteins ,0104 chemical sciences ,HEK293 Cells ,Spectrometry, Fluorescence ,chemistry ,Microscopy, Fluorescence ,Cell culture ,betalains ,biosynthesis ,mCherry ,Glycoprotein - Abstract
We designed and engineered a dye production cassette encoding a heterologous pathway, including human tyrosine hydroxylase and Amanita muscaria 4,5-DOPA dioxygenase, for the biosynthesis of the betaxanthin family of plant and fungal pigments in mammalian cells. The system does not impair cell viability, and can be used as a non-protein reporter system to directly visualize the dynamics of gene expression by profiling absorbance or fluorescence in the supernatant of cell cultures, as well as for fluorescence labeling of individual cells. Pigment profiling can also be multiplexed with reporter proteins such as mCherry or the human model glycoprotein SEAP (secreted alkaline phosphatase). Furthermore, absorbance measurement with a smartphone camera using standard application software enables inexpensive, low-tech reporter quantification., Nucleic Acids Research, 48 (12), ISSN:1362-4962, ISSN:0301-5610
- Published
- 2020
17. From synthetic biology to human therapy: engineered mammalian cells
- Author
-
Leo Scheller and Martin Fussenegger
- Subjects
0106 biological sciences ,medicine.medical_treatment ,Cell ,Biomedical Engineering ,Cell- and Tissue-Based Therapy ,Bioengineering ,Computational biology ,Biology ,01 natural sciences ,Cell therapy ,03 medical and health sciences ,Synthetic biology ,010608 biotechnology ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Treatment options ,Immunotherapy ,3. Good health ,Cancer treatment ,medicine.anatomical_structure ,Synthetic Receptors ,Synthetic Biology ,Cellular immunotherapy ,Biotechnology - Abstract
Mammalian synthetic biology has evolved to become a key driver of biomedical innovation in the area of cell therapy. Advances in receptor engineering, immunotherapy and cell implants promise new treatment options for complex diseases. Synthetic receptors have already found applications in cellular immunotherapy for cancer treatment, and are being introduced into the field of cell implants. Here, we discuss prospects for the next generation of engineered mammalian cells for human therapy, highlighting selected recent studies.
- Published
- 2018
18. Nonimmune cells equipped with T-cell-receptor-like signaling for cancer cell ablation
- Author
-
Leo Scheller, Ryosuke Kojima, and Martin Fussenegger
- Subjects
0301 basic medicine ,Cell ,Receptors, Antigen, T-Cell ,Biology ,03 medical and health sciences ,Immune system ,Antigen ,Neoplasms ,medicine ,Humans ,Molecular Targeted Therapy ,Molecular Biology ,Cell Engineering ,HEK 293 cells ,T-cell receptor ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Recombinant Proteins ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,Cancer cell ,Signal transduction ,Signal Transduction - Abstract
The ability to engineer custom cell-contact-sensing output devices into human nonimmune cells would be useful for extending the applicability of cell-based cancer therapies and for avoiding risks associated with engineered immune cells. Here we have developed a new class of synthetic T-cell receptor-like signal-transduction device that functions efficiently in human nonimmune cells and triggers release of output molecules specifically upon sensing contact with a target cell. This device employs an interleukin signaling cascade, whose OFF/ON switching is controlled by biophysical segregation of a transmembrane signal-inhibitory protein from the sensor cell-target cell interface. We further show that designer nonimmune cells equipped with this device driving expression of a membrane-penetrator/prodrug-activating enzyme construct could specifically kill target cells in the presence of the prodrug, indicating its potential usefulness for target-cell-specific, cell-based enzyme-prodrug cancer therapy. Our study also contributes to the advancement of synthetic biology by extending available design principles to transmit extracellular information to cells.
- Published
- 2017
19. Calcium‐Mediated Liposome Fusion to Engineer Giant Lipid Vesicles with Cytosolic Proteins and Reconstituted Mammalian Proteins
- Author
-
Petra S. Dittrich, Sebastian Buchmann, Yannick R. F. Schmid, and Leo Scheller
- Subjects
liposomes ,Protein reconstitution ,membrane fusion ,Biomedical Engineering ,Artificial cells ,Cell-derived vesicles ,Liposomes ,Membrane fusion ,cell-derived vesicles ,010402 general chemistry ,01 natural sciences ,General Biochemistry, Genetics and Molecular Biology ,Biomaterials ,03 medical and health sciences ,Cytosol ,Cell surface receptor ,Humans ,Cell Engineering ,Unilamellar Liposomes ,plasma-membrane vesicles ,domains ,030304 developmental biology ,0303 health sciences ,Liposome ,Artificial cell ,Chemistry ,Vesicle ,unilamellar vesicles ,Proteins ,Lipid bilayer fusion ,protein reconstitution ,0104 chemical sciences ,HEK293 Cells ,Membrane ,Biophysics ,Calcium ,complex ,artificial cells - Abstract
Giant unilamellar lipid vesicles (GUVs) are widely used as model membrane systems and provide an excellent basis to construct artificial cells. To construct more sophisticated artificial cells, proteins-in particular membrane proteins-need to be incorporated in GUVs. However, current methods for protein reconstitution have limited throughput or are not generally applicable for all proteins because they depend on detergent solubilization. This limitation is addressed here by introducing calcium-mediated membrane fusion to transfer proteins between negatively charged GUVs and cell-derived plasma membrane vesicles (CDVs), derived from HEK293T cells overexpressing a membrane receptor protein. Fusion conditions are optimized using large unilamellar vesicles and GUVs containing phosphatidylserines and fusogenic lipids. The approach is then applied to induce lipid mixing and subsequent transfer of the overexpressed membrane receptor from CDVs into GUVs. The membrane receptor is detected by immunofluorescence on GUVs that underwent lipid mixing with CDVs. Those GUVs also exhibit esterase activity because cytosolic esterases entrapped in the CDVs are transferred during membrane fusion. Thus, content mixing is demonstrated. Using CDVs circumvents the need to purify or solubilize proteins. Moreover, calcium-mediated fusion allows transfer of lipids, water-soluble and membrane bound proteins in one step, resulting in a semi-synthetic cell., Advanced Biosystems, 4 (11), ISSN:2366-7478
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.