- NCCR MSE

Projects

Displaying results 1 to 20 out of 44
« First ‹ Previous Page 1 Page 2 Page 3 Next › Last »

Description [methodology]

We engineer synthetic cellular memory platforms that enable the reconstruction of cellular histories and can be applied as living diagnostics.

A fundamental challenge in biology is to understand how cells function and integrate complex molecular information to perform different behaviors. For example, the differentiation of a stem cell into two daughter cells with distinct identities or the transformation of a normal cell into a cancer cell. This challenge has motivated the creation of numerous technologies facilitating detailed intracellular observations at the level of DNA, RNA, protein, and metabolites. Despite the power of these approaches, they generally require destructive methods and therefore observations are limited to a few snapshots in time or select asynchronous cellular processes. One provocative solution to this is to introduce DNA writing and molecular recording platforms within cells that enable the encoding, storage, and retrieval of molecular information.

Towards the goal of continuously recording molecular events within cells, my laboratory is developing and applying Record-seq, a ‘transcriptional recording’ platform that employs CRISPR spacer acquisition from RNA to capture and convert intracellular RNAs into DNA, permanently storing transcriptional information in the DNA of living cells. The newly acquired sequences serve as transcriptional records, which are retrievable via deep sequencing and can be leverage to reconstruct cellular histories. This technology provides an entirely new mode of interrogating dynamic biological and physiological processes and opens up numerous avenues for future work in engineering cellular systems.

Clinical and immunohistochemical studies indicate that the tumor microenvironment, including stromal and immunocompetent components, plays key roles in the control of tumor progression and response to treatment. Moreover, increased recognition is given to the importance of using human cell- based models to predict the outcome of therapeutic strategies.

The goal of the project is to engineer a 3D human organotypic model of colorectal cancer including associated tumor microenvironment components, as test system for new immune-oncology agents. The further inclusion of hepatocytes in the model in a second phase of the project will enable to investigate, along with the antitumor efficacy, also the possible liver toxicity of the therapeutic agents.

This project focuses on the development of an innovative methodology for the assembly of hierarchical architectures on both solid and polymer platforms. The goal is to design multi-functional systems supporting cascade reactions in 2D networks of nano-compartments (polymersomes, liposomes and combination of thereof) by their controlled anchoring and patterning on surfaces. We will develop strategies and proof-of-principle systems with material turn-over based upon either soft-condensed matter supports (polymers) or hard, metal oxide supports. New soft–hard interfaces will be designed to provide a micro- and nanoscale environment which can modulate properties and activity.

Dimethylfumarate is an emergent pharmaceutical compound with annual sales of 4412 Mio $ in 2016. Currently, the main clinical applications are in treating psoriasis and multiple sclerosis. In addition, dimethylfumarate might be therapeutic against asthma, cancer, inflammatory bowl disease, intracerebral hemorrhage, osteoarthritis, chronic pancreatitis, retinal ischemia, and ischemic stroke.

The goal of this project is to engineer a cell-like device for targeted delivery of the electrophilic drug dimethylfumarate. In the first phase of the project we plan to identify a vesicle based enzyme reaction that is triggered by external stimuli to produce and release dimethylfumarate.

An ideal cell-based diabetes therapy in humans should consist of an autonomous core system enabling closed-loop control of D-glucose sensing and insulin release, coupled to a user-defined control interface that allows the metabolically inert L-glucose to monitor the temporal activity of the therapeutic core. In this context, L-glucose functions as a patient-centred adjuvant molecule that is not only free of hepatic side- effects, but also represents an optimal nutritional sugar for diabetic patients.

The focus of this project is the design and construction of an L-glucose-dependent control interface for future cell-based diabetes therapies and the validation of L-glucose polymersome-dependent control of therapeutic transgene activities in mice.

Our research focuses on the deployment of multiple immobilized molecular catalysts on a flow microreactor platform to convert elementary starting materials over telescoped reactions into chemically and structurally complex products. The interactions of different agents in multiple compartments on a solid-state platform result in a molecular factory.

A combination of multiple catalyst permutations and applied reaction conditions enables screening of diverse parameter spaces for the conversion of suitable starting materials. This approach aims at not only synthesizing one catalysis product from one starting material, but multiple products from the same starting material and whole compound libraries. This bio-inspired approach resembles the biosynthetic processes that are taking place in a biological cell, in which multiple metabolites are often produced from a single molecule.

Our research comprises the design and synthesis of complementary catalysts, linkers and starting materials for heterogeneous catalysis with molecular catalyst monolayers, as well as the immobilization of these catalysts in flow microreactors and conducting synthetic operation on these platforms. This project is in close collaboration with the Mayor group and IBM Research - Zurich in Rüschlikon. The silicon-based flow microreactors are being fabricated by microfabrication techniques, are scalable in the number of compartments, and allow various reaction control features such as nanoscale electrode arrays, catalyst-supporting surfaces, externally controllable micro-heaters and nanophotonic sensing sites to be implemented and to be used as reaction feedback controls.

The chemical and physical processes enabling the transformation of matter in living systems is regulated by complex feedback loops. Such tightly cross-regulated processes enable highly complex reaction cascades as well as transport and exchange mechanisms that have not yet been achieved in synthetic systems. The project „Nanopores as Solid-State Approach to Interlinked Reaction Compartments" strives to simulate isolated feedback mechanisms to investigate the underlying regulation mechanisms in detail and to identify the parameters governing the system.

To fabricate such interlinked reaction compartments, we aim at fabricating nanopore devices based on a solid-state approach using top-down fabrication techniques. Self-assembled monolayers (SAMs) of functional molecular building blocks are physically separated but remain addressable by electrical, optical or electrochemical means. The SAMs are highly oriented which enables correlations between chemical structure and electronic as well as ionic transport properties in single-molecular junctions to be studied. 

Preliminary studies will allow us to mimic a biological response and to investigate isolated feedback mechanisms in detail. Additionally, from a materials point of view, the resulting oligomers may be interesting. Their physical properties are length-dependent and, thanks to a feedback mechanism, their length-distribution may become tunable by specific parameters dictated by the system, leading to a new size-control approach with wide potential applications in material science. Modular systems can easily be expanded with additional functionalities. For example redox-­dependent chromophores that will facilitate the investigation of the systems dynamics as it can be investigated by optical microscopy.

The project is not only geared towards investigating feedback mechanisms across vesicle membranes but also towards integrating vesicles as molecular factories. With such vesicles, molecular devices enabling photo-induced charge separation across the vesicle membrane will be studied. In a later stage of the project, we envisage electrochemical interconnecting of different functionalized vesicles to build up gradients of chemical potentials. 

Molecular systems derive their functional breadth from the interplay of multiple elements. The successful cooperation of these elements is often limited to narrow windows of operation, which are often difficult to identify.

We are optimizing complex in vitro systems so they can successfully operate in these windows. For this, we develop cell free systems that allow the synthesis of multiple catalysts and other protein-based elements, and compartmentalize the synthesis in nanoliter or picoliter-sized droplets. This helps us to investigate thousands of system compositions per minute. We use this to develop design rules for multi-membered systems and prepare such droplets for analysis in a classical way (i.e., by fluorescence) and by label-free methods, such as mass-spectrometry. This way we can optimize system function for a variety of objectives, ranging from enzyme evolution to the engineering of smart systems for metabolic diseases.

Building autonomous synthetic organelles and cells with a defined function using a repertoire of functional modules (toolkit) and containing inside a minimal metabolism for survival, represents the ultimate goal of this project group.

Such complex processors will open a wide variety of possibilities ranging from environmental to medical applications. One of the most important challenges will be to provide a large repertoire of engineered and modular biomolecular-transport and -energy conversion systems for assembly of nanoreactors with diverse functionalities in lipid bilayers and block copolymers.

Initially, modules will include light-­driven proton pumps and proton-driven solute transporters in the membrane, and metabolizing enzymes inside the container. Next, more complex powering systems will be explored such as combinations of light-­driven proton pumps with sodium/proton antiporters with the objective: to energise sodium-driven solute transporters. This will significantly increase the repertoire and specificity of translocating modules.

The availability of numerous, highly specialized membrane proteins in milligram amounts offers the unique opportunity to use them as building blocks and toolkit to assemble molecular factories in the form of nanoreactors and functional surfaces using bottom-up approaches.

This project group has a strong expertise and knowledge in biochemistry, function and structure of membrane proteins. Furthermore, the group already possesses a significant number of recombinant transport proteins for different solutes such as peptides, sugars, amino acids and antibiotics that can be used as modules for engineering and assembly of nanoreactors.

This project shows true interdisciplinary, transversal research: Clinical tests are conducted with light sensitive, molecular systems in partnership with the Friedrich-Miescher Institute of our industry-partner, Novartis. Should the tests be successful, this project could enable blind people to see in black and white again and, eventually, regain their full color vision.

Retinitis pigmentosa (RP) refers to a diverse group of progressive, hereditary diseases of the retina that lead to incurable blindness and affects 2 million people worldwide. Artificial photoreceptors constructed by gene delivery of light-activated channels or pumps (functional molecular modules) to surviving cell types in the remaining retinal circuit have shown to restore photosensitivity in animal models of RP at the level of the retina and cortex as well as behaviourally.

Simply said, in a degenerated macula the first step is missing: there are no more rods and cones that can detect light and subsequently convert light into neural signal. The visual nerves however are intact. In tests with apes and dogs the genetically delivered molecular factories dock successfully with the visual nerve of the eye and are activated by light, producing certain impulses that enables blind animals to see again.

Protein engineering is the field of discovering novel, synthetic proteins or improving a known protein’s activity (e.g., antibody binding affinity for a particular antigen) by using either rational or directed evolution methods. A new method that combines elements of both rational design and directed evolution is deep mutational scanning, which combines high-throughput screening with next-generation DNA sequencing to assess the functional impact of mutations across the protein sequence.

This project aims to establish deep mutational scanning as a viable approach for engineering complex therapeutic proteins. As a starting point, we will use already existing therapeutic monoclonal antibodies against cancer targets, such as Herceptin, Avastin, Cetuximab, and Rituximab, and improve their binding affinity and expression stability. We will also use sequencing data and combine it with biophysical and structural modelling to determine if we can improve the functional scoring systems generated by deep mutational scanning.

One major objective in synthetic biology is the bottom-up assembly of functional nanocells. These structures consist of lipid or polymer membranes, which serve as architectural scaffolds for functional modules such as membrane and soluble proteins. The former is embedded in the membrane, while the latter is encapsulated inside the container. The correct orientation of membrane proteins in the membrane is essential in order to avoid functional short circuits and obtain fully functional nanocells.

In this project, we propose the use of fusion proteins combined with clipping proteins to functionalize nanocells with more than one type of membrane protein.

Molecular Systems Engineering (MSE) incarnates a novel approach to clinical innovation that considerably expands the toolbox of molecular sciences and healthcare both theoretically and technically. The prospects of this emerging field to bring about scientific and clinical innovation crucially depend on proactively addressing potential ethical and regulatory bottlenecks.

Such issues include three major domains, that is: issues associated with society’s appraisal of the novel bio-technological characteristics of engineered molecular systems; the ethical and legal aspects linked to the clinical translation of MSE into healthcare applications; and the development of appropriate regulatory standards for the assessment of MSE applications by regulatory agencies.

To address those issues, this project brings new empirical research and analytical competences on ethics and regulatory issues of MSE technologies. Thanks to considerable experience and reputation in the field of bioethics and health policy, the ETH Zürich’s Health Ethics and Policy lab led by the PIs Effy Vayena, and Alessandro Blasimme will ensure dedicated research on all of the above issues.

Normative and empirical research on the ethics of MSE will result in specific ethical guidelines to guide the long-term development of the field in the future. As far as regulatory aspects are concerned, relevant national and international stakeholders – including regulators – will be engaged and a regulatory roadmap for MSE will be developed.

This project uses state-of-the-art molecular and genomic editing platforms to engineer immune cells for applications in biotechnology and cellular immunotherapy.

Our ongoing projects are focused on applying molecular and genome editing tools to engineer various types of immune cells. The precise genomic exchange of highly similar immunogenomic genes has not been demonstrated before using genome-editing tools, thus a series of aims and milestones to advance this goal have been established. For example, in one of aims we are engineering mammalian cellular factories for protein production. In another aim we are using precise genome editing to improve cellular therapeutics for transplantation and cancer. We will combine our efforts with the network of NCCR researchers to push the boundaries of immunological systems engineering. 

Scientific Highlights

  • Plug-and-(dis)play mammalian cells. We have used precise genome editing to develop a platform for rapid generation of stable cell lines capable of surface expression and secretion of recombinant proteins. The simplicity of our plug-and-(dis)play platform is highlighted by the fact that it only requires a single transfection and screening step to generate stable cells. We envision these cell lines can be applied for generating recombinant protein reagents and therapeutics. 
  • Reprogramming MHC-specificity immune cells. We have established a proof-of-concept for MHC-allelic replacement, which could be used in the future for improving the donor-host matching, which is a major challenge in allogeneic cellular transplantations in cancer. We used genome editing methods to precisely exchange the MHC region of immune cells, which were then subsequently verified for functions immune activity. Our methods can be applied for the engineering of other immunogenomic regions, which would have value in cellular immunotherapy.

Publications

N. Ayoub, P. Roth, Z. Ucurum, D. Fotiadis, S. Hirschi “Structural and biochemical insights into His-tag-induced higher-order oligomerization of membrane proteins by cryo-EM and size exclusion chromatography“ J. Struct. Biol. 2023. [DOI]
X. Chen, F. Bayard, N. Gonzalez-Sanchis, K. K. P. Pamungkas, N. Sakai, S. Matile “Fluorescent Flippers: Small-Molecule Probes to Image Membrane Tension in Living Systems“ Angew. Chem. Int. Ed. 2023. [DOI]
M. R. Schreier, B. Pfund, D. M. Steffen, O. Wenger “Photocatalytic Regeneration of a Nicotinamide Adenine Nucleotide Mimic with Water-Soluble Iridium(III) Complexes“ Inorg. Chem. 2023. [DOI]
J. Lopez-Morales, R. Vanella, G. Kovacevic, M. S. Santos, M. Nash “Titrating Avidity of Yeast-Displayed Proteins Using a Transcriptional Regulator“ ACS Synth. Biol. 2023. [DOI]
V. Mihali, M. Skowicki, D. Messmer, C. G. Palivan “Clusters of polymersomes and Janus nanoparticles hierarchically self-organized and controlled by DNA hybridization“ Nano Today 2023. [DOI]
I. Incaviglia, S. Herzog, G. Fläschner, N. Strohmeyer, E. Tosoratti, D. J. Müller “Tailoring the Sensitivity of Microcantilevers To Monitor the Mass of Single Adherent Living Cells“ Nano Lett. 2023. [DOI]
A. Bertschi, B. Stefanov, S. Xue, G. Charpin-El Hamri, A. P. Teixeira, M. Fussenegger “Controlling therapeutic protein expression via inhalation of a butter flavor molecule“ Nucleic Acids Res. 2023. [DOI]
A. García-García, S. Pigeot, I. Martin “Engineering of immunoinstructive extracellular matrices for enhanced osteoinductivity“ Bioact. Mater. 2023. [DOI]
O. M. Eggenberger, P. Jaśko, S. Tarvirdipour, C. Schoenenberger, C. G. Palivan “Advances in biohybridized planar polymer membranes and membrane‐like matrices“ Helv. Chim. Acta 2023. [DOI]
Z. Jončev, C. Sparr “Atroposelective Arene-Forming Alkene Metathesis“ Angew. Chem. Int. Ed. 2022. [DOI]
M. Mahameed, P. Wang, S. Xue, M. Fussenegger “Engineering receptors in the secretory pathway for orthogonal signalling control“ Nat. Commun. 2022. [DOI]
G. Born, E. Plantier, G. Nannini, A. Caimi, A. Mazzoleni, M. A. Asnaghi, M. G. Muraro, A. Scherberich, I. Martin, A. García-García “Mini- and macro-scale direct perfusion bioreactors with optimized flow for engineering 3D tissues“ Biotechnol. J. 2022. [DOI]
J. L. Mehl, A. Earle, J. Lammerding, M. Mhlanga, V. Vogel, N. Jain “Blockage of lamin-A/C loss diminishes the pro-inflammatory macrophage response“ iScience 2022. [DOI]
R. Castellanos-Rueda, R. B. Di Roberto, F. Bieberich, F. S. Schlatter, D. Palianina, O. T. P. Nguyen, E. Kapetanovic, H. Läubli, A. Hierlemann, N. Khanna, S. Reddy “speedingCARs: accelerating the engineering of CAR T cells by signaling domain shuffling and single-cell sequencing“ Nat. Commun. 2022. [DOI]
T. A. Schmidt, C. Sparr “Photocatalytic deracemisation of cobalt(iii) complexes with fourfold stereogenicity“ Chem. Commun. 2022. [DOI]
P. Valmaggia, N. Inglin, P. Kaiser, H. Scholl, P. M. Maloca “Iris Color Matters—A Contractility Analysis With Dynamic Volume-Rendered Optical Coherence Tomography Pupillometry“ Transl. Vis. Sci. Technol. 2022. [DOI]
S. Galvan, O. Madderson, S. Xue, A. P. Teixeira, M. Fussenegger “Regulation of Transgene Expression by the Natural Sweetener Xylose“ Adv. Sci. 2022. [DOI]
I. Shybeka, J. R. J. Maynard, S. Saidjalolov, D. Moreau, N. Sakai, S. Matile “Dynamic Covalent Michael Acceptors to Penetrate Cells: Thiol-Mediated Uptake with Tetrel-Centered Exchange Cascades, Assisted by Halogen-Bonding Switches“ Angew. Chem. Int. Ed. 2022. [DOI]
K. M. Castro, A. Scheck, S. Xiao, B. E. Correia “Computational design of vaccine immunogens“ Curr. Opin. Biotechnol. 2022. [DOI]
M. Mansouri, P. G. Ray, N. Franko, S. Xue, M. Fussenegger “Design of programmable post-translational switch control platform for on-demand protein secretion in mammalian cells“ Nucleic Acids Res. 2022. [DOI]
J. Dong, A. Ostertag, C. Sparr “o-Quinodimethane Atropisomers: Enantioselective Synthesis and Stereospecific Transformation“ Angew. Chem. Int. Ed. 2022. [DOI]
L. Kouba, J. Bürgin, G. Born, G. Perale, D. J. Schaefer, A. Scherberich, S. Pigeot, I. Martin “A composite, off-the-shelf osteoinductive material for large, vascularized bone flap prefabrication“ Acta Biomaterialia 2022. [DOI]
J. A. Kübler, B. Pfund, O. Wenger “Zinc(II) Complexes with Triplet Charge-Transfer Excited States Enabling Energy-Transfer Catalysis, Photoinduced Electron Transfer, and Upconversion“ JACS Au 2022. [DOI]
J. S. Fleck, S. M. J. Jansen, D. Wollny, F. Zenk, M. Seimiya, A. Jain, R. Okamoto, M. Santel, Z. He, J. G. Camp, B. Treutlein “Inferring and perturbing cell fate regulomes in human brain organoids“ Nature 2022. [DOI]
S. Hirschi, T. R. Ward, W. P. Meier, D. J. MüllerD. Fotiadis “Synthetic Biology: Bottom-Up Assembly of Molecular Systems“ Chem. Rev. 2022. [DOI]
R. Vazquez-Lombardi, J. S. Jung, F. S. Schlatter, A. Mei, N. R. Mantuano, F. Bieberich, K. Hong, J. Kucharczyk, E. Kapetanovic, E. Aznauryan, C. R. Weber, A. Zippelius, H. Läubli, S. Reddy “High-throughput T cell receptor engineering by functional screening identifies candidates with enhanced potency and specificity“ Immunity 2022. [DOI]
M. S. Muthwill, P. Kong, I. A. Dinu, D. Necula, C. John, C. G. Palivan “Tailoring Polymer-Based Nanoassemblies for Stimuli-Responsive Theranostic Applications“ Macromol. Biosci. 2022. [DOI]
B. A. Stefanov, M. Fussenegger “Biomarker-driven feedback control of synthetic biology systems for next-generation personalized medicine“ Front. Bioeng. Biotechnol. 2022. [DOI]
J. Vallapurackal, A. Stucki, A. D. Liang, J. Klehr, P. DittrichT. R. Ward “Ultrahigh-Throughput Screening of an Artificial Metalloenzyme using Double Emulsions“ Angew. Chem. Int. Ed. 2022. [DOI]
B. Stefanov, M. Mansouri, G. Charpin-El Hamri, M. Fussenegger “Sunlight-Controllable Biopharmaceutical Production for Remote Emergency Supply of Directly Injectable Therapeutic Proteins“ Small 2022. [DOI]
F. Risser, J. López-Morales, M. Nash “Adhesive Virulence Factors of Staphylococcus aureus Resist Digestion by Coagulation Proteases Thrombin and Plasmin“ ACS Bio. Med. Chem. Au 2022. [DOI]
H. Chen, T. Li, N. Sakai, C. Besnard, L. Guénée, M. Pupier, J. Viger-Gravel, K. TiefenbacherS. Matile “Decoded fingerprints of hyperresponsive, expanding product space: polyether cascade cyclizations as tools to elucidate supramolecular catalysis“ Chemical Science 2022. [DOI]
M. A. Beliaeva, F. Seebeck “Discovery and Characterization of the Metallopterin-Dependent Ergothioneine Synthase from Caldithrix abyssi“ JACS Au 2022. [DOI]
T. J. Enz, P. M. Maloca, M. Tschopp, M. N. Menke, J. R. Tribble, P. A. Williams, N. Inglin, U. Steitz, H. Scholl, A. Papazoglou “Volume-rendered optical coherence tomography angiography during ocular interventions: Advocating for noninvasive intraoperative retinal perfusion monitoring“ J. Biophotonics 2022. [DOI]
T. Strittmatter, Y. Wang, A. Bertschi, L. Scheller, P. C. Freitag, P. G. Ray, P. Stuecheli, J. V. Schaefer, T. Reinberg, D. Tsakiris, A. Plückthun, H. Ye, M. Fussenegger “Programmable DARPin-based receptors for the detection of thrombotic markers“ Nat. Chem. Biol. 2022. [DOI]
P. M. Maloca, C. Freichel, C. Hänsli, P. Valmaggia, P. L. Müller, S. Zweifel, C. Seeger, N. Inglin, H. Scholl, N. Denk “Cynomolgus monkey’s choroid reference database derived from hybrid deep learning optical coherence tomography segmentation“ Scientific Reports 2022. [DOI]
J. Wang, S. Lisanza, D. Juergens, D. Tischer, J. L. Watson, K. M. Castro, R. Ragotte, A. Saragovi, L. F. Milles, M. Baek, I. Anishchenko, W. Yang, D. R. Hicks, M. Expòsit, T. Schlichthaerle, J. Chun, J. Dauparas, N. Bennett, B. I. M. Wicky, A. Muenks, F. DiMaio, B. Correia, S. Ovchinnikov, D. Baker “Scaffolding protein functional sites using deep learning“ Science 2022. [DOI]
T. Li, F. Huck, G. Piccini, K. Tiefenbacher “Mimicry of the proton wire mechanism of enzymes inside a supramolecular capsule enables β-selective O-glycosylations“ Nat. Chem. 2022. [DOI]
T. A. Duncombe, A. Ponti, P. Dittrich “SpectraSorter: Ocean Insight spectrometer software application for high-throughput full-spectrum UV–VIS analysis and triggered sorting“ SoftwareX 2022. [DOI]
M. Bina, A. Krywko-Cendrowska, D. Daubian, W. Meier, C. G. Palivan “Multicomponent Copolymer Planar Membranes with Nanoscale Domain Separation“ Nano Lett. 2022. [DOI]
S. Tarvirdipour, M. Skowicki, C. Schoenenberger, L. E. Kapinos, R. Y. H. Lim, Y. BenensonC. G. Palivan “A self-assembling peptidic platform to boost the cellular uptake and nuclear delivery of oligonucleotides“ Biomater. Sci. 2022. [DOI]
A. P. Cuny, K. Tanuj Sapra, D. Martinez-Martin, G. Fläschner, J. D. Adams, S. Martin, C. Gerber, F. Rudolf, D. J. Müller “High-resolution mass measurements of single budding yeast reveal linear growth segments“ Nat. Commun. 2022. [DOI]
V. Haellman, M. Pirkl, A. Akmammedov, P. Saxena, N. Beerenwinkel, R. Paro, A. P. Teixeira, M. Fussenegger “dCas9-mediated dysregulation of gene expression in human induced pluripotent stem cells during primitive streak differentiation“ Metab. Eng. 2022 [More Information]
M. Fussenegger “Autonomous push button-controlled rapid insulin release from a piezoelectrically activated subcutaneous cell implant“ Sci. Adv. 2022. [DOI]
N. Sinha, B. Pfund, C. Wegeberg, A. Prescimone, O. Wenger “Cobalt(III) Carbene Complex with an Electronic Excited-State Structure Similar to Cyclometalated Iridium(III) Compounds“ J. Am. Chem. Soc. 2022. [DOI]
“A cholesterol analog stabilizes the human beta2-adrenergic receptor nonlinearly with temperature“ Sci. Signal. 2022. [DOI]
A. Najer, A. Belessiotis-Richards, H. Kim, C. Saunders, F. Fenaroli, C. Adrianus, J. Che, R. L. Tonkin, H. Høgset, S. Lörcher, M. Penna, S. G. Higgins, W. Meier, I. Yarovsky, M. M. Stevens “Block Length-Dependent Protein Fouling on Poly(2-oxazoline)-Based Polymersomes: Influence on Macrophage Association and Circulation Behavior“ Small 2022. [DOI]
S. Di Leone, M. Kyropoulou, J. Köchlin, R. Wehr, W. P. Meier, C. G. Palivan “Tailoring a Solvent-Assisted Method for Solid-Supported Hybrid Lipid–Polymer Membranes“ Langmuir 2022. [DOI]
M. Korpidou, V. Maffeis, I. A. Dinu, C. Schoenenberger, W. P. Meier, C. G. Palivan “Inverting glucuronidation of hymecromone in situ by catalytic nanocompartments“ J. Mater. Chem. B 2022. [DOI]
L. Heuberger, M. Korpidou, O. M. Eggenberger, M. Kyropoulou, C. G. Palivan “Current Perspectives on Synthetic Compartments for Biomedical Applications“ Int. J. Mol. Sci. 2022. [DOI]
Z. Liu, B. Yang, J. Lopez Morales, M. Nash “Optimal Sacrificial Domains in Mechanical Polyproteins: S. epidermidis Adhesins Are Tuned for Work Dissipation“ JACS Au 2022. [DOI]
R. Platt “Noninvasive assessment of gut function using transcriptional recording sentinel cells“ Science 2022. [DOI]
S. A. Mari, K. Pluhackova, J. Pipercevic, M. Leipner, S. Hiller, A. Engel, D. J. Müller “Gasdermin-A3 pore formation propagates along variable pathways“ Nat. Commun. 2022. [DOI]
L. Assies, V. Mercier, J. López-Andarias, A. Roux, N. Sakai, S. Matile “The Dynamic Range of Acidity: Tracking Rules for the Unidirectional Penetration of Cellular Compartments“ ChemBioChem 2022. [DOI]
D. Monserrat Lopez, V. Grimaudo, G. Prone, A. Flisch, A. Riedo, R. Zboray, T. Lüthi, M. MayorM. Fussenegger, P. Broekmann, P. Wurz, E. Lörtscher “Automated, 3-D and Sub-Micron Accurate Ablation-Volume Determination by Inverse Molding and X-Ray Computed Tomography“ Adv. Sci. 2022. [DOI]
M. R. Schreier, X. Guo, B. Pfund, Y. Okamoto, T. R. Ward, C. Kerzig, O. Wenger “Water-Soluble Tris(cyclometalated) Iridium(III) Complexes for Aqueous Electron and Energy Transfer Photochemistry“ Acc. Chem. Res. 2022. [DOI]
T. Kato, B. Lim, Y. Cheng, A. Pham, J. Maynard, D. Moreau, A. I. Poblador-Bahamonde, N. Sakai, S. Matile “Cyclic Thiosulfonates for Thiol-Mediated Uptake: Cascade Exchangers, Transporters, Inhibitors“ JACS Au 2022. [DOI]
M. Mansouri, M. Fussenegger “Electrogenetics: Bridging synthetic biology and electronics to remotely control the behavior of mammalian designer cells“ Current Opinion in Chemical Biology 2022. [DOI]
A. Bharioke, M. Munz, A. Brignall, G. Kosche, M. F. Eizinger, N. Ledergerber, D. Hillier, B. Gross-Scherf, K. Conzelmann, E. Macé, B. Roska “General anesthesia globally synchronizes activity selectively in layer 5 cortical pyramidal neurons“ Neuron 2022. [DOI]
Q. Laurent, N. Sakai, S. Matile “An Orthogonal Dynamic Covalent Chemistry Tool for Ring-Opening Polymerization of Cyclic Oligochalcogenides on Detachable Helical Peptide Templates“ Chem. Eur. J. 2022. [DOI]
A. Marchand, A. K. Van Hall-Beauvais, B. Correia “Computational design of novel protein–protein interactions – An overview on methodological approaches and applications“ Curr. Opin. Struct. Biol. 2022. [DOI]
X. Kong, M. Ibrahim-Ahmed, M. G. Bittencourt, R. W. Strauss, D. G. Birch, A. V. Cideciyan, A. Ervin, A. Ho, J. S. Sunness, I. S. Audo, M. Michaelides, E. Zrenner, S. Sadda, M. S. Ip, S. West, H. Scholl “Longitudinal Changes in Scotopic and Mesopic Macular Function as Assessed with Microperimetry in Patients With Stargardt Disease: SMART Study Report No. 2“ 2022. [DOI]
B. Lim, T. Kato, C. Besnard, A. I. Poblador Bahamonde, N. Sakai, S. Matile “Pnictogen-Centered Cascade Exchangers for Thiol-Mediated Uptake: As(III)-, Sb(III)-, and Bi(III)-Expanded Cyclic Disulfides as Inhibitors of Cytosolic Delivery and Viral Entry“ JACS Au 2022. [DOI]
A. Scheck, S. Rosset, M. Defferrard, A. Loukas, J. Bonet, P. Vandergheynst, B. Correia “RosettaSurf—A surface-centric computational design approach“ PLoS Comput. Biol. 2022. [DOI]
L. Deblock, E. Goossens, R. Pokratath, K. De Buysser, J. De Roo “Mapping out the Aqueous Surface Chemistry of Metal Oxide Nanocrystals: Carboxylate, Phosphonate, and Catecholate Ligands“ JACS Au 2022. [DOI]
M. Stauffer, J. Jeckelmann, H. Ilgü, Z. Ucurum, R. Boggavarapu, D. Fotiadis “Peptide transporter structure reveals binding and action mechanism of a potent PEPT1 and PEPT2 inhibitor“ Commun. Chem. 2022. [DOI]
T. Gerber, C. Loureiro, N. Schramma, S. Chen, A. Jain, A. Weber, A. Weigert, M. Santel, K. Alim, B. Treutlein, J. G. Camp “Spatial transcriptomic and single-nucleus analysis reveals heterogeneity in a gigantic single-celled syncytium“ eLife 2022. [DOI]
X. Wu, C. Sparr “Retrosynthetic polyketide disconnections for unnatural aromatics“ Trends Chem. 2022. [DOI]
X. Wu, C. Sparr “Stereoselective Synthesis of Atropisomeric Acridinium Salts by the Catalyst-Controlled Cyclization of ortho-Quinone Methide Iminiums“ Angew. Chem. Int. Ed. 2022. [DOI]
J. García-Calvo, J. López-Andarias, J. Maillard, V. Mercier, C. Roffay, A. Roux, A. Fürstenberg, N. Sakai, S. Matile “HydroFlipper membrane tension probes: imaging membrane hydration and mechanical compression simultaneously in living cells“ Chem. Sci. 2022. [DOI]
R. Vanella, G. Kovacevic, V. Doffini, J. Fernández de Santaella, M. Nash “High-throughput screening, next generation sequencing and machine learning: advanced methods in enzyme engineering“ Chem. Commun. 2022. [DOI]
M. Mahameed, M. Fussenegger “Engineering Autonomous Closed-loop Designer Cells for Disease Therapy“ iScience 2022. [DOI]
A. Stein, A. D. Liang, R. Sahin, T. R. Ward “Incorporation of metal-chelating unnatural amino acids into halotag for allylic deamination“ J. Organomet. Chem. 2022. [DOI]
M. Mahameed, S. Xue, B. Stefanov, G. C. Hamri, M. Fussenegger “Engineering a Rapid Insulin Release System Controlled By Oral Drug Administration“ Adv. Sci. 2022. [DOI]
H. Wang, M. Xie, G. Rizzi, X. Li, K. Tan, M. Fussenegger “Identification of Sclareol As a Natural Neuroprotective Cav1.3-Antagonist Using Synthetic Parkinson-Mimetic Gene Circuits and Computer-Aided Drug Discovery“ Adv. Sci. 2022. [DOI]
H. Liu, R. A. Moreira, A. Dujmović, B. Yang, A. B. Poma, M. Nash “Mapping Mechanostable Pulling Geometries of a Therapeutic Anticalin/CTLA-4 Protein Complex“ Nano Lett. 2022. [DOI]
P. Dittrich, N. Nuti, P. Rottmann, A. Stucki, P. Koch, S. Panke “A Multiplexed Cell-Free Assay to Screen for Antimicrobial Peptides in Double Emulsion Droplets“ Angew. Chem. Int. Ed. 2022. [DOI]
A. P. Teixeira, P. Stücheli, D. Ausländer, P. Schönenberger, S. Hürlemann, M. Fussenegger “CelloSelect – A synthetic cellobiose metabolic pathway for selection of stable transgenic CHO cell lines“ Metab. Eng. 2022. [DOI]
Z. He, A. Maynard, A. Jain, T. Gerber, R. Petri, H. Lin, M. Santel, K. Ly, J. Dupré, L. Sidow, F. Sanchis Calleja, S. M. J. Jansen, S. Riesenberg, J. G. Camp, B. Treutlein “Lineage recording in human cerebral organoids“ Nat. Methods 2022. [DOI]
L. Syntrivanis, K. Tiefenbacher “Catalysis Inside the Hexameric Resorcinarene Capsule: Toward Addressing Current Challenges in Synthetic Organic Chemistry“ Supramolecular Catalysis 2022. [DOI]
X. Huang, D. Hürlimann, H. T. Spanke, D. Wu, M. Skowicki, I. A. Dinu, E. R. Dufresne, C. G. Palivan “Cell-Derived Vesicles with Increased Stability and On-Demand Functionality by Equipping Their Membrane with a Cross-Linkable Copolymer“ Advanced Healthcare Materials 2022. [DOI]
X. Wu, J. Malinčík, A. Prescimone, C. Sparr “X-Ray Crystallographic Studies of Quasi-Racemates for Absolute Configuration Determinations“ Helv. Chim. Acta 2022. [DOI]
H. Chen, A. Frontera, M. . Gutiérrez López, N. Sakai, S. Matile “Pnictogen-Bonding Catalysts, Compared to Tetrel-Bonding Catalysts:  More Than Just Weak Lewis Acids“ Helvetica Chimica Acta 2022. [DOI]
L. Prochazka, Y. S. Michaels, C. Lau, R. D. Jones, M. Siu, T. Yin, D. Wu, E. Jang, M. Vázquez-Cantú, P. M. Gilbert, H. Kaul, Y. Benenson, P. W. Zandstra “Synthetic gene circuits for cell state detection and protein tuning in human pluripotent stem cells“ Mol. Syst. Biol. 2022. [DOI]
D. Moser, C. Sparr “Synthesis of Atropisomeric Two-Axis Systems by the Catalyst-Controlled syn- and anti-Selective Arene-Forming Aldol Condensation“ Angew. Chem. Int. Ed. 2022. [DOI]
G. Prone, U. Drechsler, C. Mayr, O. Semenova, C. Thanner, A. Olziersky, M. Eibelhuber, M. MayorE. Lörtscher “Scalable, Nanometer-Accurate Fabrication of All-Dielectric Metasurfaces with Narrow Resonances Tunable from Near Infrared to Visible Wavelengths“ Adv. Photonics Res. 2022. [DOI]
N. Sakai, L. Assies, S. Matile “G-Quartets, 4-Way Junctions and Triple Helices but Not DNA Duplexes: Planarization of Twisted Push-Pull Flipper Probes by Surface Recognition Rather Than Physical Compression“ Helv. Chim. Acta 2022. [DOI]
F. Ruggeri, C. Schwemmer, M. Stauffer, P. M. Nicollier, J. Figueiredo da Silva, P. D. Bosshart, K. Kochems, D. Fotiadis, A. Knoll, H. Wolf “Placement of Biological Membrane Patches in a Nanofluidic Gap With Control Over Position and Orientation“ 2022. [DOI]
N. R. Weisbach, A. Meijs, R. Platt “Multiplexed Genome Engineering with Cas12a“ 2022. [DOI]
M. Y. Cherepkova, T. Tanna, R. Platt “Recording Biological Information with CRISPR - Cas Systems“ Crispr 2022. [DOI]
M. Mansouri, M. Fussenegger “Remote Control of Mammalian Therapeutic Designer Cells“ Cell Engineering 2021. [DOI]
P. R. Laskowski, K. Pluhackova, M. Haase, B. M. Lang, G. Nagler, A. Kuhn, D. J. Müller “Monitoring the binding and insertion of a single transmembrane protein by an insertase“ Nat. Commun. 2021. [DOI]
L. Assies, J. García-Calvo, F. Piazzolla, S. Sanchez, T. Kato, L. Reymond, A. Goujon, A. Colom, J. López-Andarias, K. Straková, D. Mahecic, V. Mercier, M. Riggi, N. Jiménez-Rojo, C. Roffay, G. Licari, M. Tsemperouli, F. Neuhaus, A. Fürstenberg, E. Vauthey, S. Hoogendoorn, M. Gonzalez-Gaitan, A. Zumbuehl, K. Sugihara, J. Gruenberg, H. Riezman, R. Loewith, S. Manley, A. Roux, N. Winssinger, N. Sakai, S. Pitsch, S. Matile “Flipper Probes for the Community“ Chimia 2021. [DOI]
J. García-Calvo, J. López-Andarias, N. Sakai, S. Matile “Planarizable Push-Pull Probes with Sulfoximine-Bridged Dithienothiophene Acceptors“ Helv. Chim. Acta 2021. [DOI]
Y. Benenson “An AAV gene therapy computes over multiple cellular inputs to enable precise targeting of multifocal hepatocellular carcinoma in mice“ Sci. Transl. Med. 2021. [DOI]
R. Altamura, J. Doshi, Y. Benenson “Rational design and construction of multi-copy biomanufacturing islands in mammalian cells“ Nucleic Acids Res. 2021. [DOI]
C. Miró-Vinyals, A. Stein, S. Fischer, T. R. Ward, A. Deliz Liang “HaloTag Engineering for Enhanced Fluorogenicity and Kinetics with a Styrylpyridium Dye“ ChemBioChem 2021. [DOI]
N. Ritzmann, S. Manioglu, S. Hiller, D. J. Müller “Monitoring the antibiotic darobactin modulating the β-barrel assembly factor BamA“ Structure 2021. [DOI]
M. Kuhn, A. J. Santinha, R. Platt “Moving from in vitro to in vivo CRISPR screens“ Gene and Genome Editing 2021. [DOI]
M. Parvizian, J. De Roo “Precursor chemistry of metal nitride nanocrystals“ Nanoscale 2021. [DOI]
A. Stein, D. Chen, N. V. Igareta, Y. Cotelle, J. G. Rebelein, T. R. Ward “A Dual Anchoring Strategy for the Directed Evolution of Improved Artificial Transfer Hydrogenases Based on Carbonic Anhydrase“ ACS Cent. Sci. 2021. [DOI]
N. Romantini, S. Alam, S. Dobitz, M. Spillmann, M. De Foresta, R. Schibli, G. F. X. Schertler, H. Wennemers, X. Deupi, M. Behe, P. Berger “Exploring the signaling space of a GPCR using bivalent ligands with a rigid oligoproline backbone“ Proc. Natl. Acad. Sci. U.S.A. 2021. [DOI]
M. S. Santos, H. Liu, V. Schittny, R. Vanella, M. Nash “Correlating Single-molecule Rupture Mechanics with Cell Population Adhesion by Yeast Display of Monomeric Streptavidin“ Biophys. Rep. 2021. [DOI]
N. Franko, A. P. Teixeira, S. Xue, G. Charpin-El Hamri, M. Fussenegger “Design of modular autoproteolytic gene switches responsive to anti-coronavirus drug candidates“ Nat. Commun. 2021. [DOI]
M. Calcabrini, D. Van den Eynden, S. S. Ribot, R. Pokratath, J. Llorca, J. De Roo, M. Ibáñez “Ligand Conversion in Nanocrystal Synthesis: The Oxidation of Alkylamines to Fatty Acids by Nitrate“ JACS Au 2021. [DOI]
L. Zartner, V. Maffeis, C. Schoenenberger, I. A. Dinu, C. G. Palivan “Membrane protein channels equipped with a cleavable linker for inducing catalysis inside nanocompartments“ -11-2021 J. Mater. Chem. B 2021. [DOI]
J. López-Andarias, K. Eblighatian, Q. T. L. Pasquer, L. Assies, N. Sakai, S. Hoogendoorn, S. Matile “Photocleavable Fluorescent Membrane Tension Probes: Fast Release with Spatiotemporal Control in Inner Leaflets of Plasma Membrane, Nuclear Envelope, and Secretory Pathway“ Angew. Chem. Int. Ed. 2021. [DOI]
A. García-García, I. Martin “Biomimetic human bone marrow tissues: models to study hematopoiesis and platforms for drug testing“ Mol. Cell. Oncol. 2021. [DOI]
E. Klingler, U. Tomasello, J. Prados, J. M. Kebschull, A. Contestabile, G. L. Galiñanes, S. Fièvre, A. Santinha, R. Platt, D. Huber, A. Dayer, C. Bellone, D. Jabaudon “Temporal controls over inter-areal cortical projection neuron fate diversity“ Nature 2021. [DOI]
R. Martinent, S. Tawffik, J. López-Andarias, D. Moreau, Q. Laurent, S. Matile “Dithiolane quartets: thiol-mediated uptake enables cytosolic delivery in deep tissue“ Chem. Sci. 2021. [DOI]
M. Mansouri, M. Fussenegger “Synthetic Biology-based Optogenetic Approaches to Control Therapeutic Designer Cells“ Curr. Opin. Syst. Biol. 2021. [DOI]
A. García-García, T. Klein, G. Born, M. Hilpert, A. Scherberich, C. Lengerke, R. C. Skoda, P. E. Bourgine, I. Martin “Culturing patient-derived malignant hematopoietic stem cells in engineered and fully humanized 3D niches“ Proc. Natl. Acad. Sci. U.S.A. 2021. [DOI]
N. Meral, O. Zabek, H. Camenzind Zuche, U. Müller, D. Prétot, A. Rickmann, H. Scholl, M. della Volpe Waizel “Metabolic long-term monitoring of transcorneal electrical stimulation in retinitis pigmentosa“ Ophthalmic Res. 2021. [DOI]
S. Shui, P. Gainza, L. Scheller, C. Yang, Y. Kurumida, S. Rosset, S. Georgeon, R. B. Di Roberto, R. Castellanos-Rueda, S. ReddyB. Correia “A rational blueprint for the design of chemically-controlled protein switches“ Nat. Commun. 2021. [DOI]
M. Mansouri, M. Fussenegger “Therapeutic cell engineering: designing programmable synthetic genetic circuits in mammalian cells“ Protein Cell 2021. [DOI]
G. Born, M. Nikolova, A. Scherberich, B. Treutlein, A. García-García, I. Martin “Engineering of fully humanized and vascularized 3D bone marrow niches sustaining undifferentiated human cord blood hematopoietic stem and progenitor cells“ J. Tissue Eng. 2021. [DOI]
H. V. Humeniuk, A. Gini, X. Hao, F. Coelho, N. Sakai, S. Matile, M. Fussenegger “Pnictogen-Bonding Catalysis and Transport Combined: Polyether Transporters Made In Situ“ JACS Au 2021. [DOI]
A. Viljoen, M. Mathelié-Guinlet, A. Ray, N. Strohmeyer, Y. J. Oh, P. Hinterdorfer, D. J. Müller, D. Alsteens, Y. F. Dufrêne “Force spectroscopy of single cells using atomic force microscopy“ Nat. Rev. Methods Primers 2021. [DOI]
T. A. Duncombe, A. Ponti, F. SeebeckP. Dittrich “UV–Vis Spectra-Activated Droplet Sorting for Label-Free Chemical Identification and Collection of Droplets“ Anal. Chem. 2021. [DOI]
B. Turpin, E. Y. Bijman, H. Kaltenbach, J. Stelling “Population Design for Synthetic Gene Circuits“ Computational Methods in Systems Biology 2021. [DOI]
C. E. Meyer, C. Schoenenberger, R. P. Wehr, D. Wu, C. G. Palivan “Artificial Melanogenesis by Confining Melanin/Polydopamine Production inside Polymersomes“ Macromol. Biosci. 2021. [DOI]
T. Strittmatter, P. Argast, P. Buchman, K. Krawczyk, M. Fussenegger “Control of gene expression in engineered mammalian cells with a programmable shear-stress inducer“ Biotechnol. Bioeng. 2021. [DOI]
B. Stefanov, A. P. Teixeira, M. Mansouri, A. Bertschi, K. Krawczyk, G. C. Hamri, S. Xue, M. Fussenegger “Genetically Encoded Protein Thermometer Enables Precise Electrothermal Control of Transgene Expression“ Adv. Sci. 2021. [DOI]
R. Wehr, E. C. dos Santos, M. S. Muthwill, V. Chimisso, J. Gaitzsch, W. Meier “Fully amorphous atactic and isotactic block copolymers and their self-assembly into nano- and microscopic vesicles“ Polym. Chem. 2021. [DOI]
A. Baiyoumy, J. Vallapurackal, F. Schwizer, T. Heinisch, T. Kardashliev, M. Held, S. PankeT. R. Ward “Directed Evolution of a Surface-Displayed Artificial Allylic Deallylase Relying on a GFP Reporter Protein“ ACS Catal. 2021. [DOI]
B. Lozhkin, T. R. Ward “Bioorthogonal strategies for the in vivo synthesis or release of drugs“ Bioorg. Med. Chem. 2021. [DOI]
L. Acevedo Rua, M. Mumme, C. Manferdini, S. Darwiche, A. Khalil, M. Hilpert, D. A. Buchner, G. Lisignoli, P. Occhetta, B. von Rechenberg, M. Haug, D. J. Schaefer, M. Jakob, A. Caplan, I. Martin, A. Barbero, K. Pelttari “Engineered nasal cartilage for the repair of osteoarthritic knee cartilage defects“ Sci. Transl. Med. 2021. [DOI]
V. Maffeis, A. Belluati, I. Craciun, D. Wu, S. Novak, C. Schoenenberger, C. G. Palivan “Clustering of catalytic nanocompartments for enhancing an extracellular non-native cascade reaction“ Chem. Sci. 2021. [DOI]
M. Stauffer, Z. Ucurum, D. Harder, D. Fotiadis “Engineering and functional characterization of a proton-driven β-lactam antibiotic translocation module for bionanotechnological applications“ Sci. Rep. 2021. [DOI]
S. J. Nemat, K. Tiefenbacher “Thioderivatives of Resorcin[4]arene and Pyrogallol[4]arene: Are Thiols Tolerated in the Self-Assembly Process?“ Org. Lett. 2021. [DOI]
S. Tarvirdipour, M. Skowicki, C. Schoenenberger, C. G. Palivan “Peptide-Assisted Nucleic Acid Delivery Systems on the Rise“ Int. J. Mol. Sci. 2021. [DOI]
T. A. Schmidt, C. Sparr “Catalyst-Controlled Stereoselective Barton–Kellogg Olefination“ Angew. Chem. Int. Ed. 2021. [DOI]
H. Lin, Z. He, S. Ebert, M. Schörnig, M. Santel, M. T. Nikolova, A. Weigert, W. Hevers, N. N. Kasri, E. Taverna, J. G. Camp, B. Treutlein “NGN2 induces diverse neuron types from human pluripotency“ Stem Cell Rep. 2021. [DOI]
A. Azizoglu, R. Brent, F. Rudolf “A precisely adjustable, variation-suppressed eukaryotic transcriptional controller to enable genetic discovery“ eLife 2021. [DOI]
F. Christoffel, N. V. Igareta, M. M. Pellizzoni, L. Tiessler-Sala, B. Lozhkin, D. C. Spiess, A. Lledós, J. Maréchal, R. L. Peterson, T. R. Ward “Design and evolution of chimeric streptavidin for protein-enabled dual gold catalysis“ Nat. Catal. 2021. [DOI]
B. Lim, Y. Cheng, T. Kato, A. Pham, E. Le Du, A. K. Mishra, E. Grinhagena, D. Moreau, N. Sakai, J. Waser, S. Matile “Inhibition of Thiol-Mediated Uptake with Irreversible Covalent Inhibitors“ Helv. Chim. Acta 2021. [DOI]
S. Hirschi, D. Kalbermatter, Z. Ucurum, T. Lemmin, D. Fotiadis “Cryo-EM structure and dynamics of the green-light absorbing proteorhodopsin“ Nat. Commun. 2021. [DOI]
M. Mansouri, S. Xue, M. Hussherr, T. Strittmatter, G. Camenisch, M. Fussenegger “Smartphone-Flashlight-Mediated Remote Control of Rapid Insulin Secretion Restores Glucose Homeostasis in Experimental Type-1 Diabetes“ Small 2021. [DOI]
O. Madderson, A. P. Teixeira, M. Fussenegger “Emerging mammalian gene switches for controlling implantable cell therapies“ Curr. Opin. Chem. Biol. 2021. [DOI]
Q. Laurent, R. Martinent, D. Moreau, N. Winssinger, N. Sakai, S. Matile “Oligonucleotide Phosphorothioates Enter Cells by Thiol-Mediated Uptake“ 2021. [DOI]
K. Jahnke, N. Ritzmann, J. Fichtler, A. Nitschke, Y. Dreher, T. Abele, G. Hofhaus, I. Platzman, R. R. Schröder, D. J. Müller, J. P. Spatz, K. Göpfrich “Proton gradients from light-harvesting E. coli control DNA assemblies for synthetic cells“ Nat. Commun. 2021. [DOI]
A. Stucki, P. Jusková, N. Nuti, S. Schmitt, P. Dittrich “Synchronized Reagent Delivery in Double Emulsions for Triggering Chemical Reactions and Gene Expression“ Small Methods 2021. [DOI]
S. Di Leone, J. Vallapurackal, S. Yorulmaz Avsar, M. Kyropolou, T. R. WardC. G. Palivan, W. Meier “Expanding the Potential of the Solvent-Assisted Method to Create Bio-Interfaces from Amphiphilic Block Copolymers“ Biomacromolecules 2021. [DOI]
M. Mansouri, M. Hussherr, T. Strittmatter, P. Buchmann, S. Xue, G. Camenisch, M. Fussenegger “Smart-watch-programmed green-light-operated percutaneous control of therapeutic transgenes“ Nat. Commun. 2021. [DOI]
C. Brunner, M. Grillet, A. Urban, B. Roska, G. Montaldo, E. Macé “Whole-brain functional ultrasound imaging in awake head-fixed mice“ Nat. Protoc. 2021. [DOI]
X. Hao, T. Li, H. Chen, A. Gini, X. Zhang, S. Rosset, C. Mazet, K. TiefenbacherS. Matile “Bioinspired Ether Cyclizations within a π-Basic Capsule Compared to Autocatalysis on π-Acidic Surfaces and Pnictogen-Bonding Catalysts“ Chem. Eur. J. 2021. [DOI]
D. Scherrer, D. Vogel, U. Drechsler, A. Olziersky, C. SparrM. MayorE. Lörtscher “Monitoring Solid-phase Reactions in Self-assembled Monolayers by Surface-enhanced Raman Spectroscopy“ Angew. Chem. Int. Ed. 2021. [DOI]
J. Sahel, E. Boulanger-Scemama, C. Pagot, A. Arleo, F. Galluppi, J. N. Martel, S. D. Esposti, A. Delaux, J. de Saint Aubert, C. de Montleau, E. Gutman, I. Audo, J. Duebel, S. Picaud, D. Dalkara, L. Blouin, M. Taiel, B. Roska “Partial recovery of visual function in a blind patient after optogenetic therapy“ Nat. Med. 2021. [DOI]
Q. Yu, U. Kilik, E. M. Holloway, Y. Tsai, C. Harmel, J. H. Wu, M. Czerwinski, C. J. Childs, Z. He, M. M. Capeling, S. Huang, I. A. Glass, P. D. R. Higgins, B. Treutlein, J. R. Spence, J. G. Camp “Charting human development using a multi-endodermal organ atlas and organoid models“ Cell 2021. [DOI]
G. Fläschner, C. I. Roman, N. Strohmeyer, D. Martinez-Martin, D. J. Müller “Rheology of rounded mammalian cells over continuous high-frequencies“ Nat. Commun. 2021. [DOI]
K. Bian, C. Gerber, A. J. Heinrich, D. J. Müller, S. Scheuring, Y. Jiang “Scanning probe microscopy“ Nat. Rev. Methods Primers 2021. [DOI]
M. Della Volpe Waizel, H. Scholl, M. G. Todorova “Microvascular and Metabolic Alterations in Retinitis Pigmentosa and Stargardt Disease“ Acta Ophthalmol. 2021. [DOI]
S. Fischer, T. R. Ward, A. D. Liang “Engineering a Metathesis-Catalyzing Artificial Metalloenzyme Based on Halotag“ ACS Catal. 2021. [DOI]
S. J. Nemat, D. Van den Eynden, L. Deblock, M. Heilmann, J. M. Köster, M. Parvizian, K. TiefenbacherJ. De Roo “Resorcin[4]Arene-Based Multidentate Phosphate Ligands with Superior Binding Affinity for Nanocrystal Surfaces“ Chem. Commun. 2021. [DOI]
Q. Laurent, R. Martinent, B. Lim, A. Pham, T. Kato, J. López-Andarias, N. Sakai, S. Matile “Thiol-Mediated Uptake“ JACS Au 2021. [DOI]
C. E. Meyer, C. Schoenenberger, J. Liu, I. Craciun, C. G. Palivan “DNA-Tethered Polymersome Clusters as Nanotheranostic Platform“ Chimia 2021. [DOI]
V. Haellman, T. Strittmatter, A. Bertschi, P. Stücheli, M. Fussenegger “A Versatile Plasmid Architecture for Mammalian Synthetic Biology (Vamsyb)“ Metab. Eng. 2021. [DOI]
F. Sedlmayer, A. Woischnig, V. Unterreiner, F. Fuchs, D. Baeschlin, N. Khanna, M. Fussenegger “5-Fluorouracil Blocks Quorum-Sensing of Biofilm-Embedded Methicillin-Resistant Staphylococcus Aureus in Mice“ Nucleic Acids Res. 2021. [DOI]
D. M. Mason, S. Friedensohn, C. R. Weber, C. Jordi, B. Wagner, S. M. Meng, R. A. Ehling, L. Bonati, J. Dahinden, P. Gainza, B. CorreiaS. Reddy “Optimization of Therapeutic Antibodies by Predicting Antigen Specificity from Antibody Sequence Via Deep Learning“ Nat. Biomed. Eng. 2021. [DOI]
M. Kyropoulou, S. Yorulmaz Avsar, C. Schoenenberger, C. G. Palivan, W. P. Meier “From spherical compartments to polymer films: exploiting vesicle fusion to generate solid supported thin polymer membranes“ Nanoscale 2021. [DOI]
M. Heilmann, M. Knezevic, G. Piccini, K. Tiefenbacher “Understanding the Binding Properties of Phosphorylated Glycoluril-Derived Molecular Tweezers and Selective Nanomolar Binding of Natural Polyamines in Aqueous Solution“ Org. Biomol. Chem. 2021. [DOI]
C. Lormeau, F. Rudolf, J. Stelling “A Rationally Engineered Decoder of Transient Intracellular Signals“ Nat. Commun. 2021. [DOI]
J. García-Calvo, J. López-Andarias, N. Sakai, S. Matile “The Primary Dipole of Flipper Probes“ Chem. Commun. 2021. [DOI]
V. Haellman, P. Saxena, Y. Jiang, M. Fussenegger “Rational Design and Optimization of Synthetic Gene Switches for Controlling Cell-Fate Decisions in Pluripotent Stem Cells“ Metab. Eng. 2021. [DOI]
T. Strittmatter, S. Egli, A. Bertschi, R. Plieninger, D. Bojar, M. Xie, M. Fussenegger “Gene switch for l-glucose-induced biopharmaceutical production in mammalian cells“ Biotechnol. Bioeng. 2021. [DOI]
S. Bachler, M. Ort, S. D. Krämer, P. Dittrich “Permeation Studies across Symmetric and Asymmetric Membranes in Microdroplet Arrays“ Anal. Chem. 2021. [DOI]
J. S. Fleck, F. Sanchís-Calleja, Z. He, M. Santel, M. J. Boyle, J. G. Camp, B. Treutlein “Resolving Organoid Brain Region Identities by Mapping Single-Cell Genomic Data to Reference Atlases“ Cell Stem Cell 2021. [DOI]
M. A. Beliaeva, F. Leisinger, F. Seebeck “In Vitro Reconstitution of a Five-Step Pathway for Bacterial Ergothioneine Catabolism“ ACS Chem. Biol. 2021. [DOI]
A. Stucki, J. Vallapurackal, T. R. WardP. Dittrich “Droplet Microfluidics and Directed Evolution of Enzymes: an Intertwined Journey“ Angew. Chem. Int. Ed. 2021. [DOI]
F. Piazzolla, V. Mercier, L. Assies, N. Sakai, A. Roux, S. Matile “Fluorescent Membrane Tension Probes for Early Endosomes“ Angew. Chem. Int. Ed. 2021. [DOI]
T. Vornholt, F. Christoffel, M. M. Pellizzoni, S. PankeT. R. Ward, M. Jeschek “Systematic Engineering of Artificial Metalloenzymes for New-to-Nature Reactions“ Sci. Adv. 2021. [DOI]
J. B. Bilger, C. Kerzig, C. B. Larsen, O. Wenger “A Photorobust Mo(0) Complex Mimicking [Os(2,2′-bipyridine)3]2+ and Its Application in Red-to-Blue Upconversion“ J. Am. Chem. Soc. 2021. [DOI]
V. Chimisso, S. Conti, P. Kong, C. Fodor, W. P. Meier “Metal Cation Responsive Anionic Microgels: Behaviour Towards Biologically Relevant Divalent and Trivalent Ions“ Soft Matter 2021. [DOI]
J. López-Andarias, K. Straková, R. Martinent, N. Jiménez-Rojo, H. Riezman, N. Sakai, S. Matile “Genetically Encoded Supramolecular Targeting of Fluorescent Membrane Tension Probes within Live Cells: Precisely Localized Controlled Release by External Chemical Stimulation“ JACS Au 2021. [DOI]
M. Reich, M. Dreesbach, D. Boehringer, J. Schottenhamml, E. Gehring, H. Scholl, N. Inglin, H. Agostini, T. Reinhard, W. A. Lagrèze, R. F. Spaide, C. Lange, P. M. Maloca “Negative Vessel Remodeling in Stargardt Disease Quantified with Volume-Rendered Optical Coherence Tomography Angiography“ Retina 2021. [DOI]
C. Yang, F. Sesterhenn, J. Bonet, E. A. van Aalen, L. Scheller, L. A. Abriata, J. T. Cramer, X. Wen, S. Rosset, S. Georgeon, T. Jardetzky, T. Krey, M. Fussenegger, M. Merkx, B. Correia “Bottom-up de novo design of functional proteins with complex structural features“ Nat. Chem. Biol. 2021. [DOI]
S. Merget, L. Catti, S. Zev, D. T. Major, N. Trapp, K. Tiefenbacher “Concentration-dependent Self-assembly of an Unusually Large Hexameric Hydrogen-bonded Molecular Cage“ Chem. Eur. J. 2020. [DOI]
S. Bachler, D. Haidas, M. Ort, T. A. Duncombe, P. Dittrich “Microfluidic platform enables tailored translocation and reaction cascades in nanoliter droplet networks“ Commun. Biol. 2020. [DOI]
F. Zuttion, A. Colom, S. Matile, D. Farago, F. Pompeo, J. Kokavecz, A. Galinier, J. Sturgis, I. Casuso “High-speed atomic force microscopy highlights new molecular mechanism of daptomycin action“ Nat. Commun. 2020. [DOI]
B. Yang, Z. Liu, R. Doenen, M. Nash “Influence of Fluorination on Single-Molecule Unfolding and Rupture Pathways of a Mechanostable Protein Adhesion Complex“ Nano Lett. 2020. [DOI]
D. Daubian, A. Fillion, J. Gaitzsch, W. Meier “One-Pot Synthesis of an Amphiphilic ABC Triblock Copolymer PEO-b-PEHOx-b-PEtOz and Its Self-Assembly into Nanoscopic Asymmetric Polymersomes“ Macromolecules 2020. [DOI]
R. B. Di Roberto, R. Castellanos-Rueda, S. Frey, D. Egli, R. Vazquez-Lombardi, E. Kapetanovic, J. Kucharczyk, S. Reddy “A Functional Screening Strategy for Engineering Chimeric Antigen Receptors with Reduced on-Target, Off-Tumor Activation“ Mol. Ther. 2020. [DOI]
J. Doshi, K. Willis, A. Madurga, C. Stelzer, Y. Benenson “Multiple Alternative Promoters and Alternative Splicing Enable Universal Transcription-Based Logic Computation in Mammalian Cells“ Cell Rep. 2020. [DOI]
F. Leisinger, D. A. Miarzlou, F. Seebeck “Non-coordinative binding of O2 at the active center of a copper-dependent enzyme“ Angew. Chem. Int. Ed. 2020. [DOI]
F. Teng, M. Fussenegger “Shedding Light on Extracellular Vesicle Biogenesis and Bioengineering“ Adv. Sci. 2020. [DOI]
M. Xie, M. Viviani, M. Fussenegger “Engineering Precision Therapies: Lessons and Motivations from the Clinic“ Synth. Biol. 2020. [DOI]
Y. Cheng, A. Pham, T. Kato, B. Lim, D. Moreau, J. López-Andarias, L. Zong, N. Sakai, S. Matile “Inhibitors of thiol-mediated uptake“ Chem. Sci. 2020. [DOI]
S. F. Berlanda, M. Breitfeld, C. L. Dietsche, P. Dittrich “Recent Advances in Microfluidic Technology for Bioanalysis and Diagnostics“ Anal. Chem. 2020. [DOI]
D. J. Müller, A. C. Dumitru, C. Lo Giudice, H. E. Gaub, P. Hinterdorfer, G. Hummer, J. J. De Yoreo, Y. F. Dufrêne, D. Alsteens “Atomic Force Microscopy-Based Force Spectroscopy and Multiparametric Imaging of Biomolecular and Cellular Systems“ Chem. Rev. 2020. [DOI]
M. Paraja, A. Gini, N. Sakai, S. Matile “Pnictogen-Bonding Catalysis: An Interactive Tool to Uncover Unorthodox Mechanisms in Polyether Cascade Cyclizations“ Chem. Eur. J. 2020. [DOI]
O. Wenger “Photoactive nickel complexes in cross coupling catalysis“ Chem. Eur. J. 2020. [DOI]
E. C. dos Santos, A. Belluati, D. Necula, D. Scherrer, C. E. Meyer, R. P. Wehr, E. LörtscherC. G. Palivan, W. Meier “Combinatorial Strategy for Studying Biochemical Pathways in Double Emulsion Templated Cell-Sized Compartments“ Adv. Mater. 2020. [DOI]
Y. R. F. Schmid, L. Scheller, S. Buchmann, P. Dittrich “Calcium-Mediated Liposome Fusion to Engineer Giant Lipid Vesicles with Cytosolic Proteins and Reconstituted Mammalian Proteins“ Adv. Biosys. 2020. [DOI]
A. J. Lomakin, C. J. Cattin, D. Cuvelier, Z. Alraies, M. Molina, G. P. F. Nader, N. Srivastava, P. J. Sáez, J. M. Garcia-Arcos, I. Y. Zhitnyak, A. Bhargava, M. K. Driscoll, E. S. Welf, R. Fiolka, R. J. Petrie, N. S. De Silva, J. M. González-Granado, N. Manel, A. M. Lennon-Duménil, D. J. Müller, M. Piel “The nucleus acts as a ruler tailoring cell responses to spatial constraints“ Science 2020. [DOI]
D. Ausländer, P. F. Lang, M. Kemi, M. Fussenegger “Design of Multipartite Transcription Factors for Multiplexed Logic Genome Integration Control in Mammalian Cells“ ACS Synth. Biol. 2020. [DOI]
S. Matile “Mechanosensitive Fluorescent Probes, Changing Color Like Lobsters during Cooking: Cascade Switching Variations“ Bull. Chem. Soc. Jpn. 2020. [DOI]
O. Zabek, H. Camenzind Zuche, U. Müller, H. Scholl, A. Rickmann, M. della Volpe Waizel “Optical coherence tomography angiography findings in patients undergoing transcorneal electrical stimulation for treating retinitis pigmentosa“ Graefes Arch. Clin. Exp. Ophthalmol. 2020. [DOI]
I. Shybeka, A. Aster, Y. Cheng, N. Sakai, A. Frontera, E. Vauthey, S. Matile “Naphthalenediimides with Cyclic Oligochalcogenides in Their Core“ Chem. Eur. J. 2020. [DOI]
D. Moser, A. Castrogiovanni, D. Lotter, R. M. Witzig, V. C. Fäseke, F. C. Raps, C. Sparr “Catalytic Cascade Reactions Inspired by Polyketide Biosynthesis“ Chimia 2020. [DOI]
M. A. Beliaeva, R. Burn, D. Lim, F. Seebeck “In vitro production of ergothioneine isotopologues“ Angew. Chem. Int. Ed. 2020. [DOI]
R. Martinent, D. Du, J. López-Andarias, N. Sakai, S. Matile “Oligomers of Cyclic Oligochalcogenides for Enhanced Cellular Uptake“ ChemBioChem 2020. [DOI]
G. Brancati, B. Treutlein, J. G. Camp “Resolving Neurodevelopmental and Vision Disorders Using Organoid Single-Cell Multi-omics“ Neuron 2020. [DOI]
A. Belluati, I. Craciun, C. G. Palivan “Bioactive Catalytic Nanocompartments Integrated into Cell Physiology and Their Amplification of a Native Signaling Cascade“ ACS Nano 2020. [DOI]
C. S. Cowan, M. Renner, M. De Gennaro, B. Gross-Scherf, D. Goldblum, Y. Hou, M. Munz, T. M. Rodrigues, J. Krol, T. Szikra, R. Cuttat, A. Waldt, P. Papasaikas, R. Diggelmann, C. P. Patino-Alvarez, P. Galliker, S. E. Spirig, D. Pavlinic, N. Gerber-Hollbach, S. Schuierer, A. Srdanovic, M. Balogh, R. Panero, A. Kusnyerik, A. Szabo, M. B. Stadler, S. Orgül, S. Picelli, P. W. Hasler, A. Hierlemann, H. Scholl, G. Roma, F. Nigsch, B. Roska “Cell Types of the Human Retina and Its Organoids at Single-Cell Resolution“ Cell 2020. [DOI]
M. G. Todorova, H. Scholl, M. della Volpe-Waizell “The impact of macular edema on microvascular and metabolic alterations in retinitis pigmentosa“ Graefes Arch. Clin. Exp. Ophthalmol. 2020. [DOI]
A. Samanta, V. Sabatino, T. R. Ward, A. Walther “Functional and morphological adaptation in DNA protocells via signal processing prompted by artificial metalloenzymes“ Nat. Nanotechnol. 2020. [DOI]
K. Straková, J. López-Andarias, N. Jiménez-Rojo, J. E. Chambers, S. J. Marciniak, H. Riezman, N. Sakai, S. Matile “HaloFlippers: A General Tool for the Fluorescence Imaging of Precisely Localized Membrane Tension Changes in Living Cells“ ACS Cent. Sci. 2020. [DOI]
R. Martinent, J. López-Andarias, D. Moreau, Y. Cheng, N. Sakai, S. Matile “Automated high-content imaging for cellular uptake, from the Schmuck cation to the latest cyclic oligochalcogenides“ Beilstein J. Org. Chem. 2020. [DOI]
T. Tanna, R. Ramachanderan, R. Platt “Engineered bacteria to report gut function: technologies and implementation“ Curr. Opin. Microbiol. 2020. [DOI]
I. Némethová, L. Syntrivanis, K. Tiefenbacher “Molecular Capsule Catalysis: Ready to Address Current Challenges in Synthetic Organic Chemistry?“ Chimia 2020. [DOI]
D. Lim, X. Wen, F. Seebeck “Selenoimidazolium Salts as Supramolecular Reagents for Protein Alkylation“ ChemBioChem 2020. [DOI]
L. Zartner, M. S. Muthwill, I. A. Dinu, C. Schoenenberger, C. G. Palivan “The Rise of Bio-Inspired Polymer Compartments Responding to Pathology-Related Signals“ J. Mater. Chem. B 2020. [DOI]
M. R. Schreier, B. Pfund, X. Guo, O. Wenger “Photo-triggered hydrogen atom transfer from an iridium hydride complex to unactivated olefins“ Chem. Sci. 2020. [DOI]
M. A. Tahir, Z. P. Guven, L. R. Arriaga, B. Tinao, Y. S. Yang, A. Bekdemir, J. T. Martin, A. N. Bhanji, D. Irvine, F. Stellacci, A. Alexander-Katz “Calcium-triggered fusion of lipid membranes is enabled by amphiphilic nanoparticles“ Proc. Natl. Acad. Sci. U.S.A. 2020. [DOI]
L. Zartner, M. Garni, I. Craciun, T. Einfalt, C. G. Palivan “How Can Giant Plasma Membrane Vesicles Serve as a Cellular Model for Controlled Transfer of Nanoparticles?“ Biomacromolecules 2020. [DOI]
M. Stauffer, S. Hirschi, Z. Ucurum, D. Harder, R. Schlesinger, D. Fotiadis “Engineering and Production of the Light-Driven Proton Pump Bacteriorhodopsin in 2D Crystals for Basic Research and Applied Technologies“ Methods Protoc. 2020. [DOI]
S. J. Nemat, H. Jędrzejewska, A. Prescimone, A. Szumna, K. Tiefenbacher “Catechol[4]arene: The Missing Chiral Member of the Calix[4]arene Family“ Org. Lett. 2020. [DOI]
J. García-Calvo, J. Maillard, I. Fureraj, K. Strakova, A. Colom, V. Mercier, A. Roux, E. Vauthey, N. Sakai, A. Fürstenberg, S. Matile “Fluorescent Membrane Tension Probes for Super-Resolution Microscopy: Combining Mechanosensitive Cascade Switching with Dynamic-Covalent Ketone Chemistry“ J. Am. Chem. Soc. 2020. [DOI]
S. Höllerer, L. Papaxanthos, A. C. Gumpinger, K. Fischer, C. Beisel, K. Borgwardt, Y. Benenson, M. Jeschek “Large-Scale DNA-Based Phenotypic Recording and Deep Learning Enable Highly Accurate Sequence-Function Mapping“ Nat. Commun. 2020. [DOI]
F. C. Raps, V. C. Fäseke, D. Häussinger, C. Sparr “Catalyst-Controlled Transannular Polyketide Cyclization Cascades: Selective Folding of Macrocyclic Polyketides“ Angew. Chem. Int. Ed. 2020. [DOI]
M. Spillmann, L. Thurner, N. Romantini, M. Zimmermann, B. Meger, M. Behe, M. Waldhoer, G. F. X. Schertler, P. Berger “New Insights into Arrestin Recruitment to GPCRs“ Int. J. Mol. Sci. 2020. [DOI]
M. Mansouri, S. Lichtenstein, T. Strittmatter, P. Buchmann, M. Fussenegger “Construction of a Multiwell Light-Induction Platform for Traceless Control of Gene ExpressionGeneexpression in Mammalian Cells“ Photoswitching Proteins : Methods and Protocols 2020. [DOI]
J. Folini, W. Murad, F. Mehner, W. Meier, J. Gaitzsch “Updating radical ring-opening polymerisation of cyclic ketene acetals from synthesis to degradation“ Eur. Polym. J. 2020. [DOI]
A. Gini, M. Paraja, B. Galmés, C. Besnard, A. I. Poblador-Bahamonde, N. Sakai, A. Frontera, S. Matile “Pnictogen-bonding catalysis: brevetoxin-type polyether cyclizations“ Chem. Sci. 2020. [DOI]
I. A. Dinu, S. S. Saxer, W. P. Meier, U. Pieles, N. Bruns “Stabilizing Enzymes within Polymersomes by Co-Encapsulation of Trehalose“ Biomacromolecules 2020. [DOI]
L. Scheller, M. Schmollack, A. Bertschi, M. Mansouri, P. Saxena, M. Fussenegger “Phosphoregulated orthogonal signal transduction in mammalian cells“ Nat. Commun. 2020. [DOI]
A. Castrogiovanni, D. Lotter, F. R. Bissegger, C. Sparr “JoyaPhos: An Atropisomeric Teraryl Monophosphine Ligand“ Chem. Eur. J. 2020. [DOI]
R. Wehr, J. Gaitzsch, D. Daubian, C. Fodor, W. Meier “Deepening the insight into poly(butylene oxide)-block-poly(glycidol) synthesis and self-assemblies: micelles, worms and vesicles“ RSC Adv. 2020. [DOI]
H. J. Davis, D. Häussinger, T. R. Ward, Y. Okamoto “A visible-light promoted amine oxidation catalyzed by a Cp*Ir complex“ ChemCatChem 2020. [DOI]
A. Belluati, S. Thamboo, A. Najer, V. Maffeis, C. von Planta, I. Craciun, C. G. Palivan, W. Meier “Multicompartment Polymer Vesicles with Artificial Organelles for Signal-Triggered Cascade Reactions Including Cytoskeleton Formation“ Adv. Funct. Mater. 2020. [DOI]
V. Sabatino, D. Staub, T. R. Ward “Synthesis of N-Substituted Indoles via Aqueous Ring-Closing Metathesis“ Catal. Lett. 2020. [DOI]
D. Nelidova, R. K. Morikawa, C. S. Cowan, Z. Raics, D. Goldblum, H. Scholl, T. Szikra, A. Szabo, D. Hillier, B. Roska “Restoring light sensitivity using tunable near-infrared sensors“ Science 2020. [DOI]
J. K. Leman, B. D. Weitzner, S. M. Lewis, J. Adolf-Bryfogle, N. Alam, R. F. Alford, M. Aprahamian, D. Baker, K. A. Barlow, P. Barth, B. Basanta, B. J. Bender, K. Blacklock, J. Bonet, S. E. Boyken, P. Bradley, C. Bystroff, P. Conway, S. Cooper, B. Correia, B. Coventry, R. Das, R. M. De Jong, F. DiMaio, L. Dsilva, R. Dunbrack, A. S. Ford, B. Frenz, D. Y. Fu, C. Geniesse, L. Goldschmidt, R. Gowthaman, J. J. Gray, D. Gront, S. Guffy, S. Horowitz, P. Huang, T. Huber, T. M. Jacobs, J. R. Jeliazkov, D. K. Johnson, K. Kappel, J. Karanicolas, H. Khakzad, K. R. Khar, S. D. Khare, F. Khatib, A. Khramushin, I. C. King, R. Kleffner, B. Koepnick, T. Kortemme, G. Kuenze, B. Kuhlman, D. Kuroda, J. W. Labonte, J. K. Lai, G. Lapidoth, A. Leaver-Fay, S. Lindert, T. Linsky, N. London, J. H. Lubin, S. Lyskov, J. Maguire, L. Malmström, E. Marcos, O. Marcu, N. A. Marze, J. Meiler, R. Moretti, V. K. Mulligan, S. Nerli, C. Norn, S. Ó’Conchúir, N. Ollikainen, S. Ovchinnikov, M. S. Pacella, X. Pan, H. Park, R. E. Pavlovicz, M. Pethe, B. G. Pierce, K. B. Pilla, B. Raveh, P. D. Renfrew, S. S. R. Burman, A. Rubenstein, M. F. Sauer, A. Scheck, W. Schief, O. Schueler-Furman, Y. Sedan, A. M. Sevy, N. G. Sgourakis, L. Shi, J. B. Siegel, D. Silva, S. Smith, Y. Song, A. Stein, M. Szegedy, F. D. Teets, S. B. Thyme, R. Y. Wang, A. Watkins, L. Zimmerman, R. Bonneau “Macromolecular modeling and design in Rosetta: recent methods and frameworks“ Nat. Methods 2020. [DOI]
K. Krawczyk, S. Xue, P. Buchmann, G. Charpin-El-Hamri, P. Saxena, M. Hussherr, J. Shao, H. Ye, M. Xie, M. Fussenegger “Electrogenetic cellular insulin release for real-time glycemic control in type 1 diabetic mice“ Science 2020. [DOI]
J. Serrano-Plana, C. Rumo, J. G. Rebelein, R. L. Peterson, M. Barnet, T. R. Ward “Enantioselective Hydroxylation of Benzylic C(sp3)–H Bonds by an Artificial Iron Hydroxylase Based on the Biotin–Streptavidin Technology“ J. Am. Chem. Soc. 2020. [DOI]
M. Knezevic, M. Heilmann, G. Piccini, K. Tiefenbacher “Overriding Intrinsic Reactivity in Aliphatic C–H Oxidation: Preferential C3/C4 Oxidation of Aliphatic Ammonium Substrates“ Angew. Chem. Int. Ed. 2020. [DOI]
M. A. S. Espinosa, P. Dittrich “In-Droplet Electrophoretic Separation and Enrichment of Biomolecules“ Anal. Chem. 2020. [DOI]
G. Licari, K. Strakova, S. Matile, E. Tajkhorshid “Twisting and tilting of a mechanosensitive molecular probe detects order in membranes“ Chem. Sci. 2020. [DOI]
P. Stücheli, S. Sieber, D. W. Fuchs, L. Scheller, T. Strittmatter, P. Saxena, K. Gademann, M. Fussenegger “Genetically encoded betaxanthin-based small-molecular fluorescent reporter for mammalian cells“ Nucleic Acids Res. 2020. [DOI]
B. Pfund, D. M. Steffen, M. R. Schreier, M. Bertrams, C. Ye, K. Börjesson, O. Wenger, C. Kerzig “UV Light Generation and Challenging Photoreactions Enabled by Upconversion in Water“ J. Am. Chem. Soc. 2020. [DOI]
C. E. Meyer, S. Abram, I. Craciun, C. G. Palivan “Biomolecule–polymer hybrid compartments: combining the best of both worlds“ Phys. Chem. Chem. Phys. 2020. [DOI]
E. C. dos Santos, A. Angelini, D. Hürlimann, W. Meier, C. G. Palivan “Giant Polymer Compartments for Confined Reactions“ Chemistry 2020. [DOI]
S. Di Leone, S. Yorulmaz Avsar, A. Belluati, R. Wehr, C. G. Palivan, W. P. Meier “Polymer-Lipid Hybrid Membranes as a Model Platform to Drive Membrane-Cytochrome c Interaction and Peroxidase-like Activity“ J. Phys. Chem. B 2020. [DOI]
C. Ineichen, N. Biller-Andorno, A. Deplazes-Zemp “Between fascination and concern: an exploratory study of senior citizens’ attitudes towards synthetic biology and agricultural biotechnology“ Univ. Access Inf. Soc. 2020. [DOI]
J. Liu, I. Craciun, A. Belluati, D. Wu, S. Sieber, T. Einfalt, D. Witzigmann, M. Chami, J. Huwyler, C. G. Palivan “DNA-Directed Arrangement of Soft Synthetic Compartments and Their Behavior in Vitro and in Vivo“ Nanoscale 2020. [DOI]
F. R. Bissegger, M. Neuburger, K. Tiefenbacher “Scope of the Direct Trimerisation to C3-Symmetric Cyclotribenzylene Derivatives“ Supramol. Chem. 2020. [DOI]
S. Wäscher, A. Deplazes-Zemp “Freedom of research in a democratic society“ EMBO Rep. 2020. [DOI]
A. Krywko-Cendrowska, S. di Leone, M. Bina, S. Yorulmaz-Avsar, C. G. Palivan, W. Meier “Recent Advances in Hybrid Biomimetic Polymer-Based Films: from Assembly to Applications“ Polymers 2020. [DOI]
A. Pham, S. Matile “Peptide Stapling with Anion-π Catalysts“ Chem. Asian J. 2020. [DOI]
M. Adelaide Asnaghi, L. J. Power, A. Barbero, M. Haug, R. Köppl, D. Wendt, I. Martin “Biomarker signatures of quality for engineering nasal chondrocyte-derived cartilage“ Front. Bioeng. Biotechnol. 2020. [DOI]
S. Wäscher, N. Biller-Andorno, A. Deplazes-Zemp ““I Don’t Want to Do Anything Bad.” Perspectives on Scientific Responsibility: Results from a Qualitative Interview Study with Senior Scientists“ NanoEthics 2020. [DOI]
L. Syntrivanis, I. Némethová, D. Schmid, S. Levi, A. Prescimone, F. Bissegger, D. T. Major, K. Tiefenbacher “Four-Step Access to the Sesquiterpene Natural Product Presilphiperfolan-1β-ol and Unnatural Derivatives via Supramolecular Catalysis“ J. Am. Chem. Soc. 2020. [DOI]
M. Paraja, X. Hao, S. Matile “Polyether Natural Product Inspired Cascade Cyclizations: Autocatalysis on π-Acidic Aromatic Surfaces“ Angew. Chem. Int. Ed. 2020. [DOI]
X. Guo, Y. Okamoto, M. R. Schreier, T. R. WardO. Wenger “Reductive Amination and Enantioselective Amine Synthesis by Photoredox Catalysis“ Eur. J. Org. Chem. 2020. [DOI]
S. Hirschi, D. Kalbermatter, Z. Ucurum, D. Fotiadis “Cryo-electron microscopic and X-ray crystallographic analysis of the light-driven proton pump proteorhodopsin reveals a pentameric assembly“ J. Struct. Biol. X 2020. [DOI]
C. E. Meyer, J. Liu, I. Craciun, D. Wu, H. Wang, M. Xie, M. FusseneggerC. G. Palivan “Segregated Nanocompartments Containing Therapeutic Enzymes and Imaging Compounds within DNA-Zipped Polymersome Clusters for Advanced Nanotheranostic Platform“ Small 2020. [DOI]
S. Rigo, M. Kyropoulou, C. Schoenenberger, C. G. Palivan “Battling Bacteria with Free and Surface-Immobilized Polymeric Nanostructures“ Racing for the Surface: Pathogenesis of Implant Infection and Advanced Antimicrobial Strategies 2020. [DOI]
S. Hirschi, D. Fotiadis “Purification of Membrane Proteins by Affinity Chromatography with On-Column Protease Cleavage“ Expression, Purification, and Structural Biology of Membrane Proteins 2020. [DOI]
T. Weinert, V. Panneels “Membrane Protein Preparation for Serial Crystallography Using High-Viscosity Injectors: Rhodopsin as an Example“ Expression, Purification, and Structural Biology of Membrane Proteins 2020. [DOI]
J. López-Andarias, J. Saarbach, D. Moreau, Y. Cheng, E. Derivery, Q. Laurent, M. González-Gaitán, N. Winssinger, N. Sakai, S. Matile “Cell-Penetrating Streptavidin: A General Tool for Bifunctional Delivery with Spatiotemporal Control, Mediated by Transport Systems Such as Adaptive Benzopolysulfane Networks“ J. Am. Chem. Soc. 2020. [DOI]
M. Paraja, S. Matile “Primary Anion–π Catalysis of Epoxide-Opening Ether Cyclization into Rings of Different Sizes: Access to New Reactivity“ Angew. Chem. Int. Ed. 2020. [DOI]
S. Merget, L. Catti, G. Piccini, K. Tiefenbacher “Requirements for Terpene Cyclizations inside the Supramolecular Resorcinarene Capsule: Bound Water and Its Protonation Determine the Catalytic Activity“ J. Am. Chem. Soc. 2020. [DOI]
D. Daubian, J. Gaitzsch, W. Meier “Synthesis and complex self-assembly of amphiphilic block copolymers with a branched hydrophobic poly(2-oxazoline) into multicompartment micelles, pseudo-vesicles and yolk/shell nanoparticles“ Polym. Chem. 2020. [DOI]
B. Zilate, C. Fischer, C. Sparr “Design and application of aminoacridinium organophotoredox catalysts“ Chem. Commun. 2020. [DOI]
D. Haidas, M. Napiorkowska, S. Schmitt, P. Dittrich “Parallel Sampling of Nanoliter Droplet Arrays for Noninvasive Protein Analysis in Discrete Yeast Cultivations by MALDI-MS“ Anal. Chem. 2020. [DOI]
S. Rigo, D. Hürlimann, L. Marot, M. Malmsten, W. Meier, C. G. Palivan “Decorating Nanostructured Surfaces with Antimicrobial Peptides to Efficiently Fight Bacteria“ ACS Appl. Bio Mater. 2020. [DOI]
G. Giordano-Attianese, P. Gainza, E. Gray-Gaillard, E. Cribioli, S. Shui, S. Kim, M. Kwak, S. Vollers, A. D. J. Corria Osorio, P. Reichenbach, J. Bonet, B. Oh, M. Irving, G. Coukos, B. Correia “A computationally designed chimeric antigen receptor provides a small-molecule safety switch for T-cell therapy“ Nat. Biotechnol. 2020. [DOI]
H. V. Humeniuk, G. Licari, E. Vauthey, N. Sakai, S. Matile “Mechanosensitive Membrane Probes: Push-Pull Papillons“ Supramol. Chem. 2020. [DOI]
K. Krawczyk, L. Scheller, H. Kim, M. Fussenegger “Rewiring of endogenous signaling pathways to genomic targets for therapeutic cell reprogramming“ Nat. Commun. 2020. [DOI]
M. Rybiński, S. Möller, M. Sunnåker, C. Lormeau, J. Stelling “TopoFilter: a MATLAB package for mechanistic model identification in systems biology“ BMC Bioinf. 2020. [DOI]
C. Zelmer, L. P. Zweifel, L. E. Kapinos, I. Craciun, Z. P. Güven, C. G. Palivan, R. Y. H. Lim “Organelle-specific targeting of polymersomes into the cell nucleus“ Proc. Natl. Acad. Sci. U.S.A. 2020. [DOI]
S. Barbato, L. E. Kapinos, C. Rencurel, R. Y. Lim “Karyopherin enrichment at the nuclear pore complex attenuates Ran permeability“ J. Cell Sci. 2020. [DOI]
T. Tanna, F. Schmidt, M. Y. Cherepkova, M. Okoniewski, R. Platt “Recording transcriptional histories using Record-seq“ Nat. Protoc. 2020. [DOI]
T. Einfalt, M. Garni, D. Witzigmann, S. Sieber, N. Baltisberger, J. Huwyler, W. Meier, C. G. Palivan “Bioinspired Molecular Factories with Architecture and In Vivo Functionalities as Cell Mimics“ Adv. Sci. 2020. [DOI]
B. M. Gaub, K. C. Kasuba, E. Mace, T. Strittmatter, P. R. Laskowski, S. A. Geissler, A. Hierlemann, M. FusseneggerB. RoskaD. J. Müller “Neurons differentiate magnitude and location of mechanical stimuli“ Proc. Natl. Acad. Sci. U.S.A. 2019. [DOI]
A. Azizoglu, J. Stelling “Controlling cell-to-cell variability with synthetic gene circuits“ Biochem. Soc. Trans. 2019, 47:1795-1804. [DOI]
A. Belluati, V. Mikhalevich, S. Yorulmaz Avsar, D. Daubian, I. Craciun, M. Chami, W. P. Meier, C. G. Palivan “How do the properties of amphiphilic polymer membranes influence the functional insertion of peptide pores?“ Biomacromolecules 2019. [DOI]
A. Mazé, Y. Benenson “Artificial signaling in mammalian cells enabled by prokaryotic two-component system“ Nat. Chem. Biol. 2019. [DOI]
M. Kyropoulou, S. DiLeone, A. Lanzilotto, E. C. Constable, C. E. Housecroft, W. P. Meier, C. G. Palivan “Porphyrin Containing Polymersomes with Enhanced ROS Generation Efficiency: In Vitro Evaluation“ Macromol. Biosci. 2019. [DOI]
P. Gainza, F. Sverrisson, F. Monti, E. Rodolà, D. Boscaini, M. M. Bronstein, B. Correia “Deciphering interaction fingerprints from protein molecular surfaces using geometric deep learning“ Nat. Methods 2019. [DOI]
L. Oberhauser, S. Granziera, A. Colom, A. Goujon, V. Lavallard, S. Matile, A. Roux, T. Brun, P. Maechler “Palmitate and oleate modify membrane fluidity and kinase activities of INS-1E β-cells alongside altered metabolism-secretion coupling“ Biochim. Biophys. Acta, Mol. Cell Res. 2019. [DOI]
V. C. Fäseke, F. C. Raps, C. Sparr “Polyketide Cyclizations for the Synthesis of Polyaromatics“ Angew. Chem. Int. Ed. 2019. [DOI]
T. Brandl, C. Kerzig, L. Le Pleux, A. Prescimone, O. WengerM. Mayor “Improved Photostability of a Cu(I) Complex by Macrocyclization of the Phenanthroline Ligands“ Chem. Eur. J. 2019. [DOI]
N. Jain, T. Shahal, T. Gabrieli, N. Gilat, D. Torchinsky, Y. Michaeli, V. Vogel, Y. Ebenstein “Global modulation in DNA epigenetics during pro-inflammatory macrophage activation“ Epigenetics 2019, 14:1183-1193. [DOI]
D. Wu, S. Rigo, S. Di Leone, A. Belluati, E. C. Constable, C. E. HousecroftC. G. Palivan “Brushing the surface: cascade reactions between immobilized nanoreactors“ Nanoscale 2019. [DOI]
C. Fischer, C. Kerzig, B. Zilate, O. WengerC. Sparr “Modulation of Acridinium Organophotoredox Catalysts Guided by Photophysical Studies“ ACS Catal. 2019. [DOI]
T. Pesch, L. Bonati, W. Kelton, C. Parola, R. A. Ehling, L. Csepregi, D. Kitamura, S. Reddy “Molecular Design, Optimization, and Genomic Integration of Chimeric B Cell Receptors in Murine B Cells“ Front. Immunol. 2019. [DOI]
M. della Volpe-Waizel, H. C. Zuche, U. Müller, A. Rickmann, H. Scholl, M. G. Todorova “Metabolic monitoring of transcorneal electrical stimulation in retinitis pigmentosa“ Graefes Arch. Clin. Exp .Ophthalmol. 2019. [DOI]
X. Zhang, N. Sakai, S. Matile “Methyl Scanning for Mechanochemical Chalcogen-Bonding Cascade Switches“ ChemistryOpen 2019. [DOI]
S. Wu, Y. Zhou, D. Gerngross, M. Jeschek, T. R. Ward “Chemo-enzymatic cascades to produce cycloalkenes from bio-based resources“ Nat. Commun. 2019. [DOI]
A. Castrogiovanni, P. Herr, X. Guo, C. Larsen, C. SparrO. Wenger “Shortcuts for Electron-Transfer through the Secondary Structure of Helical Oligo-1,2-naphthylenes“ Chem. Eur. J. 2019. [DOI]
K. Strakova, A. I. Poblador-Bahamonde, N. Sakai, S. Matile “Fluorescent Flipper Probes: Comprehensive Twist Coverage“ Chem. Eur. J. 2019. [DOI]
R. M. Witzig, C. Sparr “Synthesis of Enantioenriched Tetra-ortho-3,3′-substituted Biaryls by Small-Molecule-Catalyzed Noncanonical Polyketide Cyclizations“ Synlett 2019. [DOI]
J. Folini, C. Huang, J. C. Anderson, W. P. Meier, J. Gaitzsch “Novel monomers in radical ring-opening polymerisation for biodegradable and pH responsive nanoparticles“ Polym. Chem. 2019. [DOI]
R. Schubert, S. Herzog, S. Trenholm, B. RoskaD. J. Müller “Magnetically guided virus stamping for the targeted infection of single cells or groups of cells“ Nat. Protoc. 2019. [DOI]
Q. Laurent, M. Berthet, Y. Cheng, N. Sakai, S. Barluenga, N. Winssinger, S. Matile “Probing for Thiol-Mediated Uptake into Bacteria“ ChemBioChem 2019. [DOI]
D. Bojar, M. Fussenegger “The Role of Protein Engineering in Biomedical Applications of Mammalian Synthetic Biology“ Small 2019. [DOI]
V. Chimisso, C. Fodor, W. Meier “Effect of Divalent Cation on Swelling Behavior of Anionic Microgels: Quantification and Dynamics of Ion Uptake and Release“ Langmuir 2019. [DOI]
R. M. Witzig, V. C. Fäseke, D. Häussinger, C. Sparr “Atroposelective synthesis of tetra-ortho-substituted biaryls by catalyst-controlled non-canonical polyketide cyclizations“ Nat. Catal. 2019. [DOI]
J. G. Camp, R. Platt, B. Treutlein “Mapping human cell phenotypes to genotypes with single-cell genomics“ 2019. [DOI]
A. Bornhof, M. Vázquez-Nakagawa, L. Rodríguez-Pérez, M. Ángeles Herranz, N. Sakai, N. Martín, S. Matile, J. López-Andarias “Anion–π Catalysis on Carbon Nanotubes“ Angew. Chem. Int. Ed. 2019. [DOI]
V. Chimisso, V. Maffeis, D. Hürlimann, C. G. Palivan, W. Meier “Self-Assembled Polymeric Membranes and Nanoassemblies on Surfaces: Preparation, Characterization, and Current Applications“ Macromol. Biosci. 2019. [DOI]
M. Macchione, A. Goujon, K. Strakova, H. V. Humeniuk, G. Licari, E. Tajkhorshid, N. Sakai, S. Matile “A Chalcogen-Bonding Cascade Switch for Planarizable Push–Pull Probes“ Angew. Chem. Int. Ed. 2019. [DOI]
M. Baghernejad, C. Van Dyck, J. Bergfield, D. R. Levine, A. Gubicza, J. D. Tovar, M. Calame, P. Broekmann, W. Hong “Quantum interference enhanced chemical responsivity in single-molecule dithienoborepin junctions“ Chem. Eur. J. 2019. [DOI]
D. Kaldre, F. Gallou, C. Sparr, M. Parmentier “Interface-rich Aqueous Systems for Sustainable Chemical Synthesis“ Chimia 2019, 73:714-19. [DOI]
S. Wu, Y. Zhou, J. G. Rebelein, M. Kuhn, H. Mallin, J. Zhao, N. V. Igareta, T. R. Ward “Breaking Symmetry: Engineering Single-Chain Dimeric Streptavidin as Host for Artificial Metalloenzymes“ J. Am. Chem. Soc. 2019. [DOI]
V. Sabatino, J. G. Rebelein, T. R. Ward ““Close-to-Release”: Spontaneous Bioorthogonal Uncaging Resulting from Ring-Closing Metathesis“ J. Am. Chem. Soc. 2019. [DOI]
P. Jusková, Y. R. F. Schmid, A. Stucki, S. Schmitt, M. Held, P. Dittrich ““Basicles”: Microbial Growth and Production Monitoring in Giant Lipid Vesicles“ ACS Appl. Mater. Interfaces 2019. [DOI]
D. Lim, D. Gründemann, F. Seebeck “Total Synthesis and Functional Characterization of Selenoneine“ Angew. Chem. Int. Ed. 2019. [DOI]
K. Strakova, L. Assies, A. Goujon, F. Piazzolla, H. V. Humeniuk, S. Matile “Dithienothiophenes at Work: Access to Mechanosensitive Fluorescent Probes, Chalcogen-Bonding Catalysis, and Beyond“ Chem. Rev. 2019. [DOI]
C. C. Campa, N. R. Weisbach, A. J. Santinha, D. Incarnato, R. Platt “Multiplexed Genome Engineering by Cas12a and CRISPR Arrays Encoded on Single Transcripts“ Nat. Methods 2019. [DOI]
S. Thamboo, A. Najer, A. Belluati, C. von Planta, D. Wu, I. Craciun, W. Meier, C. G. Palivan “Mimicking Cellular Signaling Pathways within Synthetic Multicompartment Vesicles with Triggered Enzyme Activity and Induced Ion Channel Recruitment“ Adv. Funct. Mater. 2019. [DOI]
J. Jüttner, A. Szabo, B. Gross-Scherf, R. K. Morikawa, S. B. Rompani, P. Hantz, T. Szikra, F. Esposti, C. S. Cowan, A. Bharioke, C. P. Patino-Alvarez, . Keles, A. Kusnyerik, T. Azoulay, D. Hartl, A. R. Krebs, D. Schübeler, R. I. Hajdu, A. Lukats, J. Nemeth, Z. Z. Nagy, R. Wu, L. Xiang, X. Fang, Z. Jin, D. Goldblum, P. W. Hasler, H. Scholl, J. Krol, B. Roska “Targeting neuronal and glial cell types with synthetic promoter AAVs in mice, non-human primates and humans“ Nat. Neurosci. 2019. [DOI]
R. W. Strauss, X. Kong, A. Ho, A. Jha, S. West, M. Ip, P. S. Bernstein, D. G. Birch, A. V. Cideciyan, M. Michaelides, J. Sahel, J. S. Sunness, E. I. Traboulsi, E. Zrenner, S. Pitetta, D. Jenkins, A. H. Hariri, S. Sadda, H. Scholl “Progression of Stargardt Disease as Determined by Fundus Autofluorescence Over a 12-Month Period: ProgStar Report No. 11“ JAMA Ophthalmol. 2019. [DOI]
M. Heilmann, K. Tiefenbacher “A Modular Phosphorylated Glycoluril-Derived Molecular Tweezer for Potent Binding of Aliphatic Diamines“ Chem. Eur. J. 2019. [DOI]
M. della Volpe-Waizel, G. L. Traber, P. Maloca, M. Zinkernagel, U. Schmidt-Erfurth, G. Rubin, B. Roska, T. Otto, R. G. Weleber, H. Scholl “New Technologies for Outcome Measures in Retinal Disease: Review from the European Vision Institute Special Interest Focus Group“ Ophthalmic Res. 2019. [DOI]
I. Martin, J. Malda, N. C. Rivron “Organs by Design: Can Bioprinting Meet Self-Organization?“ Curr. Opin. Organ. Transplant. 2019. [DOI]
H. J. Davis, T. R. Ward “Artificial Metalloenzymes: Challenges and Opportunities“ ACS Cent. Sci. 2019, 5(7):1120-36. [DOI]
N. Varma, E. Mutt, J. Mühle, V. Panneels, A. Terakita, X. Deupi, P. Nogly, G. F. X. Schertler, E. Lesca “Crystal structure of jumping spider rhodopsin-1 as a light sensitive GPCR“ Proc. Natl. Acad. Sci. U.S.A. 2019, 116(29):14547-56. [DOI]
P. Bai, Y. Liu, S. Xue, G. C. Hamri, P. Saxena, H. Ye, M. Xie, M. Fussenegger “A fully human transgene switch to regulate therapeutic protein production by cooling sensation“ Nat. Med. 2019. [DOI]
Z. P. Guven, P. H. J. Silva, Z. Luo, U. B. Cendrowska, M. Gasbarri, S. T. Jones, F. Stellacci “Synthesis and Characterization of Amphiphilic Gold Nanoparticles“ J. Vis. Exp. 2019. [DOI]
H. Liu, V. Schittny, M. Nash “Removal of a Conserved Disulfide Bond Does Not Compromise Mechanical Stability of a VHH Antibody Complex“ Nano Lett. 2019. [DOI]
Q. Zhang, K. Tiefenbacher “Sesquiterpene Cyclizations inside the Hexameric Resorcinarene Capsule: Total Synthesis of δ-Selinene and Mechanistic Studies“ Angew. Chem. Int. Ed. 2019. [DOI]
D. A. Miarzlou, F. Leisinger, D. Joss, D. Häussinger, F. Seebeck “Structure of Formylglycine-Generating Enzyme in Complex with Copper and a Substrate Reveals an Acidic Pocket for Binding and Activation of Molecular Oxygen“ Chem. Sci. 2019. [DOI]
A. Maurer, F. Leisinger, D. Lim, F. Seebeck “Structure and Mechanism of Ergothionase from Treponema denticola“ Chem. Eur. J. 2019. [DOI]
H. Liu, D. T. Ta, M. Nash “Mechanical Polyprotein Assembly Using Sfp and Sortase-Mediated Domain Oligomerization for Single-Molecule Studies“ Small Methods 2019. [DOI]
T. A. Duncombe, P. Dittrich “Droplet Barcoding: Tracking Mobile Micro-Reactors for High-Throughput Biology“ Curr. Opin. Biotechnol. 2019, 60. [DOI]
M. Tsemperouli, E. Amstad, N. Sakai, S. Matile, K. Sugihara “Black Lipid Membranes: Challenges in Simultaneous Quantitative Characterization by Electrophysiology and Fluorescence Microscopy“ Langmuir 2019. [DOI]
Y. Cheng, L. Zong, J. López-Andarias, E. Bartolami, Y. Okamoto, T. R. Ward, N. Sakai, S. Matile “Cell-Penetrating Dynamic-Covalent Benzopolysulfane Networks“ Angew. Chem. Int. Ed. 2019. [DOI]
N. Jain, J. Moeller, V. Vogel “Mechanobiology of Macrophages: How Physical Factors Coregulate Macrophage Plasticity and Phagocytosis“ Annu. Rev. Biomed. Eng. 2019, 21(1):267-97. [DOI]
H. Chassin, M. Müller, M. Tigges, L. Scheller, M. Lang, M. Fussenegger “A modular degron library for synthetic circuits in mammalian cells“ Nat. Commun. 2019. [DOI]
R. Vanella, D. T. Ta, M. Nash “Enzyme-mediated hydrogel encapsulation of single cells for high-throughput screening and directed evolution of oxidoreductases“ Biotechnol. Bioeng. 2019. [DOI]
Q. Zhang, L. Catti, L. Syntrivanis, K. Tiefenbacher “En route to terpene natural products utilizing supramolecular cyclase mimetics“ Nat. Prod. Rep. 2019. [DOI]
R. Martinent, Q. Laurent, N. Sakai, S. Matile “Cellular Uptake Mediated by Cyclic Oligochalcogenides“ Chimia 2019. [DOI]
A. Ervin, R. W. Strauss, M. I. Ahmed, D. Birch, J. Cheetham, F. L. F. III, M. S. Ip, G. J. Jaffe, M. G. Maguire, E. M. Schönbach, S. R. Sadda, S. K. West, H. Scholl “A Workshop on Measuring the Progression of Atrophy Secondary to Stargardt Disease in the ProgStar Studies: Findings and Lessons Learned“ Transl. Vis. Sci. Technol. 2019, 8(2):16. [DOI]
J. G. Rebelein, Y. Cotelle, B. Garabedian, T. R. Ward “Chemical Optimization of Whole-Cell Transfer Hydrogenation Using Carbonic Anhydrase as Host Protein“ ACS Catal. 2019. [DOI]
G. S. Pulcu, N. S. Galenkamp, Y. Qing, G. Gasparini, E. Mikhailova, S. Matile, H. Bayley “Single-Molecule Kinetics of Growth and Degradation of Cell-Penetrating Poly(disulfide)s“ J. Am. Chem. Soc. 2019. [DOI]
L. Scheller, M. Fussenegger “From synthetic biology to human therapy: engineered mammalian cells“ Curr. Opin. Biotechnol. 2019, 58:108-16. [DOI]
H. Kim, D. Bojar, M. Fussenegger “A CRISPR/Cas9-based central processing unit to program complex logic computation in human cells“ Proc. Natl. Acad. Sci. U.S.A. 2019. [DOI]
J. Gaitzsch, S. Hirschi, S. Freimann, D. Fotiadis, W. Meier “Directed Insertion of Light-Activated Proteorhodopsin into Asymmetric Polymersomes from an ABC Block Copolymer“ Nano Lett. 2019. [DOI]
F. Tolle, P. Stücheli, M. Fussenegger “Genetic circuitry for personalized human cell therapy“ Curr. Opin. Biotechnol. 2019, 59:31-38. [DOI]
D. Ehrenberg, N. Varma, X. Deupi, M. Koyanagi, A. Terakita, G. F. X. Schertler, J. Heberle, E. Lesca “The two-photon reversible reaction of the bistable jumping spider rhodopsin-1“ Biophys. J. 2019. [DOI]
M. Jakobi, F. Gallou, C. Sparr, M. Parmentier “A General Protocol for Robust Sonogashira Reactions in Micellar Medium“ Helv. Chim. Acta 2019. [DOI]
E. Bartolami, D. Basagiannis, L. Zong, R. Martinent, Y. Okamoto, Q. Laurent, T. R. Ward, M. Gonzalez-Gaitan, N. Sakai, S. Matile “Diselenolane-Mediated Cellular Uptake: Efficient Cytosolic Delivery of Probes, Peptides, Proteins, Artificial Metalloenzymes and Protein-Coated Quantum Dots“ Chem. Eur. J. 2019. [DOI]
A. Goujon, A. Colom, K. Straková, V. Mercier, D. Mahecic, S. Manley, N. Sakai, A. Roux, S. Matile “Mechanosensitive Fluorescent Probes to Image Membrane Tension in Mitochondria, Endoplasmic Reticulum, and Lysosomes“ J. Am. Chem. Soc. 2019. [DOI]
A. D. Liang, J. Serrano-Plana, R. L. Peterson, T. R. Ward “Artificial Metalloenzymes Based on the Biotin–Streptavidin Technology: Enzymatic Cascades and Directed Evolution“ Acc. Chem. Res. 2019. [DOI]
C. Beerli, A. Yakimovich, S. Kilcher, G. V. Reynoso, G. Fläschner, D. J. Müller, H. D. Hickman, J. Mercer “Vaccinia virus hijacks EGFR signalling to enhance virus spread through rapid and directed infected cell motility“ Nat. Microbiol. 2019. [DOI]
S. Hirschi, N. Fischer, D. Kalbermatter, P. R. Laskowski, Z. Ucurum, D. J. MüllerD. Fotiadis “Design and assembly of a chemically switchable and fluorescently traceable light-driven proton pump system for bionanotechnological applications“ Sci. Rep. 2019. [DOI]
T. Serdiuk, A. Steudle, S. A. Mari, S. Manioglu, H. R. Kaback, A. Kuhn, D. J. Müller “Insertion and folding pathways of single membrane proteins guided by translocases and insertases“ Sci. Adv. 2019. [DOI]
A. Belluati, I. Craciun, C. E. Meyer, S. Rigo, C. G. Palivan “Enzymatic reactions in polymeric compartments: nanotechnology meets nature“ Curr. Opin. Biotechnol. 2019. [DOI]
B. Roska “The First Steps in Vision: Cell Types, Circuits, and Repair“ EMBO Mol. Med. 2019
M. Hellstern, M. Gantenbein, L. Le Pleux, G. Puebla-Hellmann, E. LörtscherM. Mayor “Electrochemical Multiplexing: Control over Surface Functionalization by Combining a Redox-Sensitive Alkyne Protection Group with “Click”-Chemistry“ Adv. Mater. Interf. 2019. [DOI]
D. Rudin, A. Lanzilotto, F. Bachmann, C. E. Housecroft, E. C. Constable, J. Drewe, M. Haschke, S. Krähenbühl “Non-immunological toxicological mechanisms of metamizole-associated neutropenia in HL60 cells“ Biochem. Pharmacol. 2019, 163:345-356. [DOI]
E. V. Konishcheva, D. Daubian, S. Rigo, W. P. Meier “Probing membrane asymmetry of ABC polymersomes“ Chem. Commun. 2019. [DOI]
Q. Laurent, N. Sakai, S. Matile “The Opening of 1,2-Dithiolanes and 1,2-Diselenolanes: Regioselectivity, Rearrangements, and Consequences for Poly(disulfide)s, Cellular Uptake and Pyruvate Dehydrogenase Complexes“ Helv. Chim. Acta 2019. [DOI]
R. Vanella, A. Bazin, D. T. Ta, M. Nash “Genetically encoded stimuli-responsive cytoprotective hydrogel capsules for single cells provide novel genotype-phenotype linkage“ Chem. Mater. 2019. [DOI]
L. M. Lee, M. Tsemperouli, A. I. Poblador-Bahamonde, S. Benz, N. Sakai, K. Sugihara, S. Matile “Anion Transport with Pnictogen Bonds in Direct Comparison with Chalcogen and Halogen Bonds“ J. Am. Chem. Soc. 2019. [DOI]
S. Yorulmaz Avsar, M. Kyropoulou, S. Di Leone, C. Schoenenberger, W. P. Meier, C. G. Palivan “Biomolecules Turn Self-Assembling Amphiphilic Block Co-polymer Platforms Into Biomimetic Interfaces“ Front. Chem. 2019, 6:645. [DOI]
O. Wenger “Is Iron the New Ruthenium?“ Chem. Eur. J. 2019. [DOI]
J. M. Köster, D. Häussinger, K. Tiefenbacher “Activation of Primary and Secondary Benzylic and Tertiary Alkyl (sp3)C-F Bonds Inside a Self-Assembled Molecular Container“ Front. Chem. 2019. [DOI]
R. Panatala, S. Barbato, T. Kozai, J. Luo, L. E. Kapinos, R. Y. H. Lim “Nuclear Pore Membrane Proteins Self-Assemble into Nanopores“ Biochemistry 2019. [DOI]
A. Angelini, C. Fodor, W. Yave, L. Leva, A. Car, W. Meier “pH-Triggered Membrane in Pervaporation Process“ ACS Omega 2018. [DOI]
M. Garni, R. Wehr, S. Yorulmaz Avsar, C. John, C. G. Palivan, W. Meier “Polymer membranes as templates for bio-applications ranging from artificial cells to active surfaces“ Eur. Polym. J. 2018. [DOI]
D. Huemmer, S. Bachler, M. Köhler, L. M. Blank, R. Zenobi, P. Dittrich “Microfluidic platform for multimodal analysis of enzyme secretion in nanoliter droplet arrays“ Anal. Chem. 2018. [DOI]
X. Zhang, X. Hao, L. Liu, A. Pham, J. López-Andarias, A. Frontera, N. Sakai, S. Matile “Primary Anion−π Catalysis and Autocatalysis“ J. Am. Chem. Soc. 2018, 140(51):17867-17871. [DOI]
E. Macé, G. Montaldo, S. Trenholm, C. Cowan, A. Brignall, A. Urban, B. Roska “Whole-Brain Functional Ultrasound Imaging Reveals Brain Modules for Visuomotor Integration“ Neuron 2018, 100(5):1241-1251.e7. [DOI]
M. Szponarski, F. Schwizer, T. R. Ward, K. Gademann “On-cell catalysis by surface engineering of live cells with an artificial metalloenzyme“ Commun. Chem. 2018. [DOI]
D. Bojar, T. Fuhrer, M. Fussenegger “Purity by design: Reducing impurities in bioproduction by stimulus-controlled global translational downregulation of non-product proteins“ Metab. Eng. 2018. [DOI]
K. T. Sapra, P. M. Spoerri, A. Engel, D. Alsteens, D. J. Müller “Seeing and sensing single G protein-coupled receptors by atomic force microscopy“ Curr. Opin. Cell Biol. 2018, 57:25-32. [DOI]
E. V. Konishcheva, U. E. Zhumaev, W. P. Meier “Self-Assembly of PEO-b-PCL-b-PMOXA Binary Mixtures“ Macromolecules 2018. [DOI]
M. Krieg, G. Fläschner, D. Alsteens, H. E. Gaub, W. H. Roos, G. J. L. Wuite, C. Gerber, Y. F. Dufrêne, D. J. Müller “Atomic force microscopy-based mechanobiology“ Nat. Rev. Phys. 2018. [DOI]
N. Jain, V. Vogel “Spatial confinement downsizes the inflammatory response of macrophages“ Nat. Mater. 2018. [DOI]
A. Goujon, K. Straková, N. Sakai, S. Matile “Streptavidin interfacing as a general strategy to localize fluorescent membrane tension probes in cells“ Chem. Sci. 2018. [DOI]
O. Wenger “Photoactive Complexes with Earth-Abundant Metals“ J. Am. Chem. Soc. 2018. [DOI]
S. Rigo, G. Gunkel-Grabole, W. Meier, C. G. Palivan “Surfaces with Dual Functionality through Specific Coimmobilization of Self-Assembled Polymeric Nanostructures“ 2018. [DOI]
D. Kalbermatter, N. Shrestha, N. Ader-Ebert, M. Herren, P. Moll, R. K. Plemper, K. Altmann, J. P. Langedijk, F. Gall, U. Lindenmann, R. Riedl, D. Fotiadis, P. Plattet “Primary Resistance Mechanism of the Canine Distemper Virus Fusion Protein against a Small-Molecule Membrane Fusion Inhibitor“ Virus Res. 2018. [DOI]
F. Schmidt, M. Y. Cherepkova, R. Platt “Transcriptional recording by CRISPR spacer acquisition from RNA“ Nature 2018. [DOI]
J. Zhao, J. G. Rebelein, H. Mallin, C. Trindler, M. M. Pellizzoni, T. R. Ward “Genetic Engineering of an Artificial Metalloenzyme for Transfer Hydrogenation of a Self-Immolative Substrate in Escherichia coli’s Periplasm“ J. Am. Chem. Soc. 2018. [DOI]
M. Mansouri, T. Strittmatter, M. Fussenegger “Light-Controlled Mammalian Cells and Their Therapeutic Applications in Synthetic Biology“ Adv. Sci. 2018. [DOI]
C. Fischer, C. Sparr “Synthesis of 1,5-bifunctional organolithium reagents by a double directed ortho-metalation: Direct transformation of esters into 1,8-dimethoxy-acridinium salts“ Tetrahedron 2018, 74:5486-5493. [DOI]
A. Belluati, I. Craciun, J. Liu, C. G. Palivan “Nanoscale Enzymatic Compartments in Tandem Support Cascade Reactions in Vitro“ Biomacromolecules 2018. [DOI]
Y. Zhao, Y. Cotelle, L. Liu, J. López-Andarias, A. Bornhof, M. Akamatsu, N. Sakai, S. Matile “The Emergence of Anion−π Catalysis“ Acc. Chem. Res. 2018, 51(9):2255-2263. [DOI]
Y. Liu, G. Charpin-El Hamri, H. Ye, M. Fussenegger “A synthetic free fatty acid-regulated transgene switch in mammalian cells and mice“ Nucleic Acids Res. 2018. [DOI]
A. P. Teixeira, M. Fussenegger “Engineering mammalian cells for disease diagnosis and treatment“ Curr. Opin. Biotechnol. 2018. [DOI]
M. E. Idrissi, C. E. Meyer, L. Zartner, W. Meier “Nanosensors based on polymer vesicles and planar membranes: a short review“ J. Nanobiotechnol. 2018. [DOI]
N. Sakai, S. Matile “Conjugated Polyimine Dynamers as Phase-Sensitive Membrane Probes“ J. Am. Chem. Soc. 2018. [DOI]
Q. Zhang, L. Catti, K. Tiefenbacher “Catalysis inside the Hexameric Resorcinarene Capsule“ Acc. Chem. Res. 2018. [DOI]
A. Colom, E. Derivery, S. Soleimanpour, C. Tomba, M. D. Molin, N. Sakai, M. González-Gaitán, S. Matile, A. Roux “A fluorescent membrane tension probe“ Nat. Chem. 2018. [DOI]
M. Garni, T. Einfalt, R. Goers, C. G. Palivan, W. P. Meier “Live follow-up of enzymatic reactions inside the cavities of synthetic giant unilamellar vesicles (GUVs) equipped with membrane proteins mimicking cell architecture“ ACS Synth. Biol. 2018. [DOI]
G. Gunkel-Grabole, W. Meier “Biomimetic Polymer Architectures“ Chimia 2018, 72:548-548. [DOI]
C. R. Weber, D. M. Mason, S. M. Meng, V. Greiff, W. J. Kelton, S. Reddy, C. Parola “High-throughput antibody engineering in mammalian cells by CRISPR/Cas9-mediated homology-directed mutagenesis“ Nucleic Acids Res. 2018, 46:7436-7449. [DOI]
D. T. Ta, R. Vanella, M. Nash “Bioorthogonal Elastin-like Polypeptide Scaffolds for Immunoassay Enhancement“ ACS Appl. Mater. Interfaces 2018. [DOI]
M. Grogg, D. Hilvert, M. Ebert, A. K. Beck, D. Seebach, F. Kurth, P. DittrichC. Sparr, S. Wittlin, M. Rottmann, P. Mäser “Cell Penetration, Herbicidal Activity, and in-vivo-Toxicity of Oligo-Arginine Derivatives and of Novel Guanidinium-Rich Compounds Derived from the Biopolymer Cyanophycin“ Helv. Chim. Acta 2018. [DOI]
C. Fischer, C. Sparr “Configurationally Stable Atropisomeric Acridinium Fluorophores“ Synlett 2018. [DOI]
F. Kurth, P. Dittrich, P. Walde, D. Seebach “Influence of the Membrane Dye R18 and of DMSO on Cell Penetration of Guanidinium-Rich Peptides“ Chem. Biodiversity 2018. [DOI]
Q. Zhang, J. Rinkel, B. Goldfuss, J. S. Dickschat, K. Tiefenbacher “Sesquiterpene cyclizations catalysed inside the resorcinarene capsule and application in the short synthesis of isolongifolene and isolongifolenone“ Nat. Catal. 2018. [DOI]
M. Alkan-Zambada, S. Keller, L. Martínez-Sarti, A. Prescimone, J. M. Junquera-Hernández, E. C. Constable, H. J. Bolink, M. Sessolo, E. Ortí, C. E. Housecroft “[Cu(P^P)(N^N)][PF6] compounds with bis(phosphane) and 6-alkoxy, 6-alkylthio, 6-phenyloxy and 6-phenylthio-substituted 2,2′-bipyridine ligands for light-emitting electrochemical cells“  J. Mater. Chem. C 2018. [DOI]
P. Dettinger, T. Frank, M. Etzrodt, N. Ahmed, A. Reimann, C. Trenzinger, D. Loeffler, K. D. Kokkaliaris, T. Schroeder, S. Tay “Automated Microfluidic System for Dynamic Stimulation and Tracking of Single Cells“ Anal. Chem. 2018. [DOI]
D. Bojar, M. Fussenegger “Programming mammalian gene expression with the antibiotic simocyclinone D8 and the flavonoid luteolin“ AIChE J. 2018. [DOI]
G. Puebla-Hellmann, K. Venkatesan, M. MayorE. Lörtscher “Metallic nanoparticle contacts for high-yield, ambient-stable molecular-monolayer devices“ Nature 2018. [DOI]
M. Hestericová, T. Heinisch, M. Lenz, T. R. Ward “Ferritin Encapsulation of Artificial Metalloenzymes: Engineering a Tertiary Coordination Sphere for an Artificial Transfer Hydrogenase“ Dalton Trans. 2018. [DOI]
H. C. Schmidt, X. Guo, P. U. Richard, M. Neuburger, C. G. PalivanO. Wenger “Mixed-valent molecular triple-deckers“ Angew. Chem. Int. Ed. 2018. [DOI]
C. L. Draghici, V. Mikhalevich, G. Gunkel-Grabole, J. Kowal, W. P. Meier, C. G. Palivan “Biomimetic planar Polymer Membranes decorated with Enzymes as Functional Surfaces“ Langmuir 2018. [DOI]
R. Goers, J. Thoma, N. Ritzmann, A. Di Silvestro, C. Alter, G. Gunkel-Grabole, D. FotiadisD. J. Müller, W. Meier “Optimized reconstitution of membrane proteins into synthetic membranes“ Commun. Chem. 2018. [DOI]
S. Matile “Chalcogen-Bonding Catalysis: From Neutral to Cationic Benzodiselenazole Scaffolds“ 2018. [DOI]
Y. Liu, P. Bai, A. Woischnig, G. Charpin-El Hamri, H. Ye, M. Folcher, M. Xie, N. Khanna, M. Fussenegger “Immunomimetic Designer Cells Protect Mice from MRSA Infection“ Cell 2018. [DOI]
H. V. Humeniuk, A. Rosspeintner, G. Licari, V. Kilin, L. Bonacina, E. Vauthey, N. Sakai, S. Matile “White‐Fluorescent Dual‐Emission Mechanosensitive Membrane Probes that Function by Bending Rather than Twisting“ Angew. Chem. Int. Ed. 2018. [DOI]
D. Bojar, L. Scheller, G. C. Hamri, M. Xie, M. Fussenegger “Caffeine-inducible gene switches controlling experimental diabetes“ Nat. Commun. 2018. [DOI]
A. Drinnenberg, F. Franke, R. K. Morikawa, J. Jüttner, D. Hillier, P. Hantz, A. Hierlemann, R. Azeredo da Silveira, B. Roska “How Diverse Retinal Functions Arise from Feedback at the First Visual Synapse“ Neuron 2018. [DOI]
R. F. Tiefenauer, T. Dalgaty, T. Keplinger, T. Tian, C. Shih, J. Vörös, M. Aramesh “Monolayer Graphene Coupled to a Flexible Plasmonic Nanograting for Ultrasensitive Strain Monitoring“ Small 2018. [DOI]
M. Xie, M. Fussenegger “Designing cell function: assembly of synthetic gene circuits for cell biology applications“ Nat. Rev. Mol. Cell Biol. 2018. [DOI]
Z. H. d. K. Master of Arts in Transdisziplinarität, R. Stutzki “Wissensorte. Eine Publikation als Ausstellung“ 2018. [DOI]
P. U. Richard, I. Craciun, J. Gaitzsch, L. Weiner, C. G. Palivan “Delivery of ROS Generating Anthraquinones Using Reduction‐responsive Peptide‐based Nanoparticles“ Helv. Chim. Acta 2018. [DOI]
J. López‐Andarias, A. Bauzá, N. Sakai, A. Frontera, S. Matile “Remote Control of Anion–π Catalysis on Fullerene‐Centered Catalytic Triads“ Angewandte Chemie International Edition 2018. [DOI]
A. Najer, C. G. Palivan, H. Beck, W. Meier “Challenges in Malaria Management and a Glimpse at Some Nanotechnological Approaches“ Infectious Diseases and Nanomedicine III 2018. [DOI]
X. Guo, Y. Okamoto, M. R. Schreier, T. R. WardO. Wenger “Enantioselective synthesis of amines by combining photoredox and enzymatic catalysis in a cyclic reaction network“ Chem. Sci. 2018. [DOI]
Y. Okamoto, R. Kojima, F. Schwizer, E. Bartolami, T. Heinisch, S. MatileM. FusseneggerT. R. Ward “A cell-penetrating artificial metalloenzyme regulates a gene switch in a designer mammalian cell“ Nat. Commun. 2018. [DOI]
A. Link, C. Sparr “Remote Central‐to‐Axial Chirality Conversion: Direct Atroposelective Ester to Biaryl Transformation“ Angew. Chem. Int. Ed. 2018. [DOI]
J. Thoma, Y. Sun, N. Ritzmann, D. J. Müller “POTRA Domains, Extracellular Lid, and Membrane Composition Modulate the Conformational Stability of the β Barrel Assembly Factor BamA“ Structure 2018. [DOI]
D. Lotter, A. Castrogiovanni, M. Neuburger, C. Sparr “Catalyst-Controlled Stereodivergent Synthesis of Atropisomeric Multiaxis Systems“ ACS Cent. Sci. 2018. [DOI]
J. M. Köster, K. Tiefenbacher “Elucidating the Importance of Hydrochloric Acid as a Cocatalyst for Resorcinarene‐Capsule‐Catalyzed Reactions“ ChemCatChem 2018. [DOI]
R. Kojima, M. Fussenegger “Engineering Whole Mammalian Cells for Target‐Cell‐Specific Invasion/Fusion“ Adv. Sci. 2018. [DOI]
K. Strakova, S. Soleimanpour, M. Diez-Castellnou, N. Sakai, S. Matile “Ganglioside‐Selective Mechanosensitive Fluorescent Membrane Probes“ Helv. Chim. Acta 2018. [DOI]
H. Liu, D. T. Ta, M. Nash “Mechanical Polyprotein Assembly Using Sfp and Sortase‐Mediated Domain Oligomerization for Single‐Molecule Studies“ Small Methods 2018. [DOI]
M. Vandenbossche, G. Gunkel-Grabole, A. Car, L. Bernard, P. Rupper, K. Maniura-Weber, M. Heuberger, G. Faccio, D. Hegemann “Near-Surface Structure of Plasma Polymer Films Affects Surface Behavior in Water and its Interaction with Proteins“ Plasma Chem. Plasma Process. 2018. [DOI]
L. Scheller, T. Strittmatter, D. Fuchs, D. Bojar, M. Fussenegger “Generalized extracellular molecule sensor platform for programming cellular behavior“ Nat. Chem. Biol. 2018. [DOI]
A. Tastanova, M. Folcher, M. Müller, G. Camenisch, A. Ponti, T. Horn, M. S. Tikhomirova, M. Fussenegger “Synthetic biology-based cellular biomedical tattoo for detection of hypercalcemia associated with cancer“ Sci. Transl. Med. 2018. [DOI]
J. Zhao, D. G. Bachmann, M. Lenz, D. G. Gillingham, T. R. Ward “An artificial metalloenzyme for carbene transfer based on a biotinylated dirhodium anchored within streptavidin“ Catal. Sci. Technol. 2018. [DOI]
M. Macchione, M. Tsemperouli, A. Goujon, A. R. Mallia, N. Sakai, K. Sugihara, S. Matile “Mechanosensitive Oligodithienothiophenes: Transmembrane Anion Transport Along Chalcogen‐Bonding Cascades“ 2018. [DOI]
J. Thoma, S. Manioglu, D. Kalbermatter, P. D. Bosshart, D. FotiadisD. J. Müller “Protein-enriched outer membrane vesicles as a native platform for outer membrane protein studies“ Commun. Biol. 2018. [DOI]
R. Kojima, D. Bojar, G. Rizzi, G. C. Hamri, M. D. El-Baba, P. Saxena, S. Ausländer, K. R. Tan, M. Fussenegger “Designer exosomes produced by implanted cells intracerebrally deliver therapeutic cargo for Parkinson’s disease treatment“ Nat. Commun. 2018. [DOI]
A. Bornhof, A. Bauzá, A. Aster, M. Pupier, A. Frontera, E. Vauthey, N. Sakai, S. Matile “Synergistic Anion–(π)n–π Catalysis on π-Stacked Foldamers“ J. Am. Chem. Soc. 2018. [DOI]
H. Hattori, E. Kaufmann, H. Miyatake-Ondozabal, R. Berg, K. Gademann “Total Synthesis of Tiacumicin A. Total Synthesis, Relay Synthesis, and Degradation Studies of Fidaxomicin (Tiacumicin B, Lipiarmycin A3)“ J. Org. Chem. 2018. [DOI]
S. G. Keller, B. Probst, T. Heinisch, R. Alberto, T. R. Ward “Photo‐Driven Hydrogen Evolution by an Artificial Hydrogenase Utilizing the Biotin‐Streptavidin Technology“ Helv. Chim. Acta 2018. [DOI]
A. Link, C. Sparr “Stereoselective arene formation“ Chem. Soc. Rev. 2018. [DOI]
S. B. Benz, A. I. Poblador‐Bahamonde, N. Low‐Ders, S. Matile “Catalysis with Pnictogen, Chalcogen, and Halogen Bonds“ Angew. Chem. Int. Ed. 2018. [DOI]
T. Einfalt, D. Witzigmann, C. Edlinger, S. Sieber, R. Goers, A. Najer, M. Spulber, O. Onaca-Fischer, J. Huwyler, C. G. Palivan “Biomimetic artificial organelles with in vitro and in vivo activity triggered by reduction in microenvironment“ Nat. Commun. 2018. [DOI]
E. Whitehead, F. Rudolf, H. Kaltenbach, J. Stelling “Automated Planning Enables Complex Protocols on Liquid-Handling Robots“ ACS Synth. Biol. 2018, 7:922-932. [DOI]
H. Mallin, T. R. Ward “Streptavidin‐Enzyme Linked Aggregates for the One‐Step Assembly and Purification of Enzyme Cascades“ ChemCatChem 2018. [DOI]
S. Rigo, C. Cai, G. Gunkel-Grabole, L. Maurizi, X. Zhang, J. Xu, C. G. Palivan “Nanoscience-Based Strategies to Engineer Antimicrobial Surfaces“ Adv. Sci. 2018. [DOI]
B. Zilate, A. Castrogiovanni, C. Sparr “Catalyst-Controlled Stereoselective Synthesis of Atropisomers“ ACS Catalysis 2018. [DOI]
G. Wang, R. D. Chow, L. Ye, C. D. Guzman, X. Dai, M. B. Dong, F. Zhang, P. A. Sharp, R. Platt, S. Chen “Mapping a functional cancer genome atlas of tumor suppressors in mouse liver using AAV-CRISPR–mediated direct in vivo screening“ Sci. Adv. 2018. [DOI]
G. Licari, J. S. Beckwith, S. Soleimanpour, S. Matile, E. Vauthey “Detecting Order and Lateral Pressure at Biomimetic Interfaces Using a Mechanosensitive Second-Harmonic-Generation Probe“ Phys. Chem. Chem. Phys. 2018. [DOI]
P. Atukorale, Z. P. Guven, A. Bekdemir, R. Carney, R. C. Van Lehn, D. S. Yun, P. H. Jacob Silva, D. Demurtas, Y. S. Yang, A. Alexander-Katz, F. Stellacci, D. J. Irvine “Structure-property relationships of amphiphilic nanoparticles that penetrate or fuse lipid membranes“ Bioconjug. Chem. 2018. [DOI]
J. Gaitzsch, P. C. Welsch, J. Folini, C. Schoenenberger, J. C. Anderson, W. Meier “Revisiting Monomer Synthesis and Radical Ring Opening Polymerization of Dimethylated MDO Towards Biodegradable Nanoparticles for Enzymes“ Eur. Polym. J. 2018. [DOI]
X. Zhang, L. Liu, J. López-Andarias, C. Wang, N. Sakai, S. Matile “Anion-π Catalysis: Focus on Nonadjacent Stereocenters“ Helv. Chim. Acta 2018. [DOI]
H. Wang, M. Xie, G. Charpin-El Hamri, H. Ye, M. Fussenegger “Treatment of chronic pain by designer cells controlled by spearmint aromatherapy“ Nat. Biomed. Eng. 2018. [DOI]
J. Shiu, L. Aires, Z. Lin, V. Vogel “Nanopillar force measurements reveal actin-cap-mediated YAP mechanotransduction“ Nat. Cell Biol. 2018. [DOI]
I. Craciun, A. S. Denes, G. Gunkel-Grabole, A. Belluati, C. G. Palivan “Surfaces decorated with polymeric nanocompartments for pH reporting“ Helv. Chim. Acta 2018. [DOI]
P. Morelli, E. Bartolami, N. Sakai, S. Matile “Glycosylated Cell-Penetrating Poly(disulfide)s: Multifunctional Cellular Uptake at High Solubility“ Helv. Chim. Acta 2018, 101(1):e1700266. [DOI]
N. Chuard, A. I. Poblador-Bahamonde, L. Zong, E. Bartolami, J. Hildebrandt, W. Weigand, N. Sakai, S. Matile “Diselenolane-mediated cellular uptake“ 2018. [DOI]
J. G. Rebelein, T. R. Ward “In vivo catalyzed new-to-nature reactions“ Curr. Opin. Biotechnol. 2018. [DOI]
E. Konishcheva, D. Daubian, J. Gaitzsch, W. Meier “Synthesis of Linear ABC Triblock Copolymers and Their Self-Assembly in Solution“ Helv. Chim. Acta 2018. [DOI]
M. M. Pellizzoni, F. Schwizer, C. W. Wood, V. Sabatino, Y. Cotelle, S. Matile, D. N. Woolfson, T. R. Ward “Chimeric Streptavidins as Host Proteins for Artificial Metalloenzymes“ ACS Catal. 2018. [DOI]
T. Heinisch, F. Schwizer, B. Garabedian, E. Csibra, M. Jeschek, J. Vallapurackal, V. B. Pinheiro, P. Marlière, S. PankeT. R. Ward “E. coli surface display of streptavidin for directed evolution of an allylic deallylase“ Chem. Sci. 2018, 9(24):5383-5388. [DOI]
M. Dastor, J. Schreiber, L. Prochazka, B. Angelici, J. Kleinert, I. Klebba, J. Doshi, L. Shen, Y. Benenson “A Workflow for In Vivo Evaluation of Candidate Inputs and Outputs for Cell Classifier Gene Circuits“ ACS Synth. Biol. 2017. [DOI]
R. Schubert, S. Trenholm, K. Balint, G. Kosche, C. S. Cowan, M. A. Mohr, M. Munz, D. Martinez-Martin, G. Fläschner, R. Newton, J. Krol, B. G. Scherf, K. Yonehara, A. Wertz, A. Ponti, A. Ghanem, D. Hillier, K. Conzelmann, D. J. MüllerB. Roska “Virus stamping for targeted single-cell infection in vitro and in vivo“ Nat. Biotechnol. 2017. [DOI]
C. Fischer, C. Sparr “Direkte Umwandlung von Estern in Heterocyclische Fluorophore“ Angew. Chem. Int. Ed. 2017. [DOI]
X. Guo, O. Wenger “Reductive Amination by Photoredox Catalysis via Polarity-Matched Hydrogen Atom Transfer“ Angew. Chem. Int. Ed. 2017. [DOI]
K. M. Carroll, A. W. Knoll, H. Wolf, U. Duerig “Explaining the Transition from Diffusion Limited to Reaction Limited Surface Assembly of Molecular Species through Spatial Variation“ Langmuir 2017. [DOI]
A. Lanzilotto, M. Kyropoulou, E. C. Constable, C. E. Housecroft, W. P. Meier, C. G. Palivan “Porphyrin-polymer nanocompartments: singlet oxygen generation and antimicrobial activity“ 2017. [DOI]
K. M. Carroll, C. Rawlings, Y. Zhang, A. W. Knoll, S. R. Marder, H. Wolf, U. Dürig “Testing the Equivalence between Spatial Averaging and Temporal Averaging in Highly Dilute Solutions“ Langmuir 2017. [DOI]
S. Ausländer, X. Pierrat, L. Hellmann, L. Rachid, M. Fussenegger “Programmable full-adder computations in communicating three-dimensional cell cultures“ Nat. Methods 2017. [DOI]
S. Arnoldini, A. Moscaroli, M. Chabria, M. Hilbert, S. Hertig, R. Schibli, M. Béhé, V. Vogel “Novel peptide probes to assess the tensional state of fibronectin fibers in cancer“ Nat. Commun. 2017. [DOI]
R. Kojima, L. Scheller, M. Fussenegger “Nonimmune cells equipped with T-cell-receptor-like signaling for cancer cell ablation“ Nat. Chem. Biol. 2017. [DOI]
J. M. Daum, O. Keles, S. J. B. Holwerda, H. Kohler, F. M. Rijli, M. Stadler, B. Roska “The formation of the light-sensing compartment of cone photoreceptors coincides with a transcriptional switch“ eLife 2017, 6:e31437. [DOI]
N. Strohmeyer, M. Bharadwaj, M. Costell, R. Fässler, D. J. Müller “Fibronectin-bound α5β1 integrins sense load and signal to reinforce adhesion in less than a second“ Nat. Mater. 2017. [DOI]
Y. Toyoda, C. J. Cattin, M. P. Stewart, I. Poser, M. Theis, T. V. Kurzchalia, F. Buchholz, A. A. Hyman, D. J. Müller “Genome-scale single-cell mechanical phenotyping reveals disease-related genes involved in mitotic rounding“ Nat. Commun. 2017, 8(1):1266. [DOI]
T. Braeuer, Q. Zhang, K. Tiefenbacher “Iminium Catalysis inside a Self-Assembled Supramolecular Capsule: Scope and Mechanistic Studies“ J. Am. Chem. Soc. 2017. [DOI]
D. T. Ta, R. Vanella, M. Nash “Magnetic separation of elastin-like polypeptide receptors for enrichment of cellular and molecular targets“ Nano Lett. 2017. [DOI]
D. Martínez-Martín, G. Fläschner, B. Gaub, S. Martin, R. Newton, C. Beerli, J. Mercer, C. Gerber, D. J. Müller “Inertial picobalance reveals fast mass fluctuations in mammalian cells“ Nature 2017, 550(7677):500-05. [DOI]
H. Chassin, B. Geering, L. Schukur, D. Ausländer, B. Lang, M. Fussenegger “Sensing and responding to allergic response cytokines through a genetically encoded circuit“ Nat. Commun. 2017. [DOI]
M. JeschekS. PankeT. R. Ward “Artificial Metalloenzymes on the Verge of New-to-Nature Metabolism“ Trends Biotechnol. 2017. [DOI]
S. Benz, J. Mareda, C. Besnard, N. Sakai, S. Matile “Catalysis with chalcogen bonds: neutral benzodiselenazole scaffolds with high-precision selenium donors of variable strength“ Chem. Sci. 2017. [DOI]
S. G. Keller, A. Pannwitz, H. Mallin, O. WengerT. R. Ward “Streptavidin as a Scaffold for Light-Induced Long-Lived Charge Separation“ Chem. Eur. J. 2017. [DOI]
R. Newton, M. Delguste, M. Koehler, A. C. Dumitru, P. R. Laskowski, D. J. Müller, D. Alsteens “Combining confocal and atomic force microscopy to quantify single-virus binding to mammalian cell surfaces“ Nat. Protoc. 2017. [DOI]
V. Haellman, M. Fussenegger “Synthetic biology – Engineering cell-based biomedical devices“ Curr. Opin. Biomed. Eng. 2017. [DOI]
J. Gaitzsch, L. Messager, E. Morecroft, W. Meier “Vesicles in Multiple Shapes: Fine-Tuning Polymersomes’ Shape and Stability by Setting Membrane Hydrophobicity“ Polymers 2017, 9(10):483. [DOI]
S. Oesterle, D. Gerngross, S. Schmitt, T. M. Roberts, S. Panke “Efficient engineering of chromosomal ribosome binding site libraries in mismatch repair proficient Escherichia coli“ Sci. Rep. 2017, 7(1):12327. [DOI]
V. Gatterdam, A. Frutiger, K. Stengele, D. Heindl, T. Lübbers, J. Vörös, C. Fattinger “Focal molography is a new method for the in situ analysis of molecular interactions in biological samples“ Nat. Nano. 2017
L. Prochazka, Y. Benenson, P. W. Zandstra “Synthetic gene circuits and cellular decision-making in human pluripotent stem cells“ Curr. Opin. Syst. Biol. 2017. [DOI]
C. Ineichen, N. Biller-Andorno, A. Deplazes-Zemp “Image of Synthetic Biology and Nanotechnology: A Survey among University Students“ Front. Genet. 2017. [DOI]
A. Pannwitz, O. Wenger “Photoinduced Electron Transfer Coupled to Donor Deprotonation and Acceptor Protonation in a Molecular Triad Mimicking Photosystem II“ J. Am. Chem. Soc. 2017. [DOI]
L. A. Buldt, O. Wenger “Chromium complexes for luminescence, solar cells, photoredox catalysis, upconversion, and phototriggered NO release“ Chem. Sci. 2017. [DOI]
J. López-Andarias, A. Frontera, S. Matile “Anion−π Catalysis on Fullerenes“ J. Am. Chem. Soc. 2017
C. B. Larsen, O. Wenger “Photoredox Catalysis with Metal Complexes Made from Earth-Abundant Elements“ Chem. Eur. J. 2017. [DOI]
H. Grimm, M. Eggel, A. Deplazes-Zemp, N. Biller-Andorno “The Road to Hell Is Paved with Good Intentions: Why Harm–Benefit Analysis and Its Emphasis on Practical Benefit Jeopardizes the Credibility of Research“ Animals 2017. [DOI]
L. Liu, Y. Cotelle, A. Bornhof, C. Besnard, N. Sakai, S. Matile “Anion–π Catalysis of Diels–Alder Reactions“ Angew. Chem. Int. Ed. 2017. [DOI]
E. V. Konishcheva, U. E. Zhumaev, M. Kratt, V. Oehri, W. Meier “Complex Self-Assembly Behavior of Bis-hydrophilic PEO-b-PCL-b-PMOXA Triblock Copolymers in Aqueous Solution“ Macromolecules 2017. [DOI]
C. Edlinger, T. Einfalt, M. Spulber, A. Car, W. Meier, C. G. Palivan “Biomimetic Strategy To Reversibly Trigger Functionality of Catalytic Nanocompartments by the Insertion of pH-Responsive Biovalves“ Nano Lett. 2017. [DOI]
E. Karamasioti, C. Lormeau, J. Stelling “Computational design of biological circuits: Putting parts into context“ Mol. Syst. Des. Eng. 2017. [DOI]
A. Larrañaga, M. Lomora, J. R. Sarasua, C. G. Palivan, A. Pandit “Polymer capsules as micro-/nanoreactors for therapeutic applications: Current strategies to control membrane permeability“ Prog. Mater. Sci. 2017. [DOI]
R. D. Chow, C. D. Guzman, G. Wang, F. Schmidt, M. W. Youngblood, L. Ye, Y. Errami, M. B. Dong, M. A. Martinez, F. Zhang, P. Renauer, K. Bilguvar, M. Gunel, P. A. Sharp, R. Platt, S. Chen “AAV-mediated direct in vivo CRISPR screen identifies functional suppressors in glioblastoma“ Nat. Neurosci. 2017. [DOI]
C. Wang, S. Matile “Anion-π Catalysts with Axial Chirality“ Chem. Eur. J. 2017. [DOI]
F. Sedlmayer, T. Jaeger, U. Jenal, M. Fussenegger “Quorum-Quenching Human Designer Cells for Closed-Loop Control of Pseudomonas aeruginosa Biofilms“ Nano Lett. 2017. [DOI]
V. Mikhalevich, I. Craciun, M. Kyropoulou, C. G. Palivan, W. Meier “Amphiphilic peptide self-assembly: Expansion to hybrid materials“ Biomacromolecules 2017. [DOI]
P. R. Laskowski, M. Pfreundschuh, M. Stauffer, Z. Ucurum, D. FotiadisD. J. Müller “High-Resolution Imaging and Multiparametric Characterization of Native Membranes by Combining Confocal Microscopy and Atomic Force Microscopy-Based Multifunctional Toolbox“ ACS Nano 2017. [DOI]
E. Derivery, E. Bartolami, S. Matile, M. Gonzalez-Gaitan “Efficient Delivery of Quantum Dots into the Cytosol of Cells Using Cell-Penetrating Poly(disulfide)s“ J. Am. Chem. Soc. 2017. [DOI]
M. Jeschek, D. Gerngross, S. Panke “Combinatorial pathway optimization for streamlined metabolic engineering“ Curr. Opin. Biotechnol. 2017, 47:142-51. [DOI]
P. Saxena, D. Bojar, H. Zulewski, M. Fussenegger “Generation of glucose-sensitive insulin-secreting beta-like cells from human embryonic stem cells by incorporating a synthetic lineage-control network“ J. Biotechnol. 2017. [DOI]
F. Schwizer, Y. Okamoto, T. Heinisch, Y. Gu, M. M. Pellizzoni, V. Lebrun, R. Reuter, V. Köhler, J. C. Lewis, T. R. Ward “Artificial Metalloenzymes: Reaction Scope and Optimization Strategies“ Chem. Rev. 2017. [DOI]
W. Aeschimann, S. Staats, S. Kammer, N. Olieric, J. Jeckelmann, D. Fotiadis, T. Netscher, G. Rimbach, M. Cascella, A. Stocker “Self-assembled α-Tocopherol Transfer Protein Nanoparticles Promote Vitamin E Delivery Across an Endothelial Barrier“ Sci. Rep. 2017. [DOI]
N. Ritzmann, J. Thoma, S. Hirschi, D. Kalbermatter, D. FotiadisD. J. Müller “Fusion Domains Guide the Oriented Insertion of Light-Driven Proton Pumps into Liposomes“ Biophys J. 2017. [DOI]
M. Xie, D. Aubel, M. Fussenegger “Closed-loop control systems – The quest for precision therapies for diabetes“ Curr. Opin. Syst. Biol. 2017. [DOI]
Y. M. Klein, A. Prescimone, E. C. Constable, C. E. Housecroft “4,2’:6’,4”- and 3,2’:6’,3”-Terpyridines: The Conflict between Well-Defined Vectorial Properties and Serendipity in the Assembly of 1D-, 2D- and 3D-Architectures“ Materials 2017. [DOI]
A. P. Teixeira, M. Fussenegger “Synthetic biology-inspired therapies for metabolic diseases“ Curr. Opin. Biotechnol. 2017. [DOI]
T. Serdiuk, S. A. Mari, D. J. Müller “Pull-and-Paste of Single Transmembrane Proteins“ Nano Lett. 2017. [DOI]
Q. Zhang, L. Catti, J. Pleiss, K. Tiefenbacher “Terpene Cyclizations inside a Supramolecular Catalyst: Leaving Group-Controlled Product Selectivity and Mechanistic Studies“ J. Am. Chem. Soc. 2017. [DOI]
C. H. Pollok, Q. Zhang, K. Tiefenbacher, C. Merten “Chirality induction from a chiral guest to the hydrogen bonding network of its hexameric resorcinarene host capsule“ ChemPhysChem 2017. [DOI]
Y. Sakiyama, R. Panatala, R. Y. H. Lim “Structural Dynamics of the Nuclear Pore Complex“ Semin. Cell Dev. Biol. 2017. [DOI]
D. Ausländer, M. Fussenegger “Synthetic Biology‐The Synthesis of Biology“ Angew. Chem. Int. Ed. 2017, 56:6396-419. [DOI]
F. Schmidt, R. Platt “Applications of CRISPR-Cas for synthetic biology and genetic recording“ Curr. Opin. Syst. Biol. 2017, 5:9-15. [DOI]
Y. Aeschi, S. Drayss-Orth, M. Valášek, F. Raps, D. Häussinger, M. Mayor “Assembly of [2]rotaxanes in water“ Eur. J. Org. Chem. 2017. [DOI]
D. Hillier, M. Fiscella, A. Drinnenberg, S. Trenholm, S. B. Rompani, Z. Raics, G. Katona, J. Juettner, A. Hierlemann, B. Rozsa, B. Roska “Causal evidence for retina-dependent and -independent visual motion computations in mouse cortex“ Nat. Neurosci. 2017. [DOI]
M. Akamatsu, N. Sakai, S. Matile “Electric-Field-Assisted Anion−π Catalysis“ J. Am. Chem. Soc. 2017, 139:6558-61. [DOI]
D. Kalbermatter, P. Chiu, J. Jeckelmann, Z. Ucurum, T. Walz, D. Fotiadis “Electron crystallography reveals that substrate release from the PTS IIC glucose transporter is coupled to a subtle conformational change“ J. Struct. Biol. 2017. [DOI]
L. A. Büldt, O. Wenger “Chromium(0), Molybdenum(0), and Tungsten(0) Isocyanide Complexes as Luminophores and Photosensitizers with Long-Lived Excited States“ Angew. Chem. Int. Ed. 2017. [DOI]
P. Orekhov, A. Bothe, H. Steinhoff, K. V. Shaitan, S. Raunser, D. Fotiadis, R. Schlesinger, J. P. Klare, M. Engelhard “Sensory Rhodopsin I and Sensory Rhodopsin II Form Trimers of Dimers in Complex with their Cognate Transducers“ Photochem. Photobiol. 2017. [DOI]
M. Pfreundschuh, D. Harder, Z. Ucurum, D. FotiadisD. J. Müller “Detecting Ligand-Binding Events and Free Energy Landscape while Imaging Membrane Receptors at Subnanometer Resolution“ Nano Lett. 2017, 17(5):3261-69. [DOI]
C. D. Ertl, F. Brunner, E. C. Constable, C. E. Housecroft “Sweetness and light: Sugar-functionalized CˆN and NˆN ligands in [Ir(CˆN)2(NˆN)]Cl complexes“ J. Organomet. Chem. 2017. [DOI]
M. Kuss-Petermann, O. Wenger “Exceptionally Long-Lived Photodriven Multi-Electron Storage without Sacrificial Reagents“ Chem. Eur. J. 2017. [DOI]
Y. OkamotoT. R. Ward “Cross-Regulation of an Artificial Metalloenzyme“ Angew. Chem. Int. Ed. 2017. [DOI]
Y. M. Klein, A. Prescimone, M. Neuburger, E. C. Constable, C. E. Housecroft “What a difference a tail makes: 2D → 2D parallel interpenetration of sheets to interpenetrated nbo networks using ditopic-4,2′:6′,4′′-terpyridine ligands“ CrystEngComm 2017. [DOI]
L. Liu, Y. Cotelle, J. Klehr, N. Sakai, T. R. WardS. Matile “Anion-π catalysis: bicyclic products with four contiguous stereogenic centers from otherwise elusive diastereospecific domino reactions on π-acidic surfaces“ Chem. Sci. 2017, 8:3770-74. [DOI]
L. Catti, A. Pöthig, K. Tiefenbacher “Host-Catalyzed Cyclodehydration–Rearrangement Cascade Reaction of Unsaturated Tertiary Alcohols“ Adv. Synth. Catal. 2017, 359:1331-38. [DOI]
A. Lanzilotto, M. Kuss-Petermann, O. Wenger, E. C. Constable, C. E. Housecroft “Homoleptic complexes of a porphyrinatozinc(ii)-2,2':6',2''-terpyridine ligand“ Photochem. Photobiol. Sci. 2017, 16:585-95. [DOI]
R. Platt, Y. Zhou, I. M. Slaymaker, A. S. Shetty, N. R. Weisbach, J. Kim, J. Sharma, M. Desai, S. Sood, H. R. Kempton, G. R. Crabtree, G. Feng, F. Zhang “Chd8 Mutation Leads to Autistic-like Behaviors and Impaired Striatal Circuits“ Cell Rep. 2017, 19:335-50. [DOI]
Y. F. Dufrene, T. Ando, R. Garcia, D. Alsteens, D. Martinez-Martin, A. Engel, C. Gerber, D. J. Muller, D. J. Müller “Imaging modes of atomic force microscopy for application in molecular and cell biology“ Nat. Nanotechnol. 2017, 12(4):295-307. [DOI]
S. Reddy “Reprogramming MHC specificity by CRISPR-Cas9-assisted cassette exchange“ Sci. Rep. 2017, 7:45775. [DOI]
S. Ausländer, M. Fussenegger “Synthetic RNA-based switches for mammalian gene expression control“ Curr. Opin. Biotechnol. 2017, 48:54-60. [DOI]
M. Kuss-Petermann, M. Orazietti, M. Neuburger, P. Hamm, O. Wenger “Intramolecular Light-Driven Accumulation of Reduction Equivalents by Proton-Coupled Electron Transfer“ J. Am. Chem. Soc. 2017. [DOI]
G. Gunkel-Grabole, C. G. Palivan, W. Meier “Nanostructured Surfaces through Immobilization of Self-Assembled Polymer Architectures Using Thiol–Ene Chemistry“ Macromol. Mater. Eng. 2017, 302:1600363. [DOI]
M. Garni, S. Thamboo, C. Schoenenberger, C. G. Palivan “Biopores/membrane proteins in synthetic polymer membranes“ Biochim. Biophys. Acta, Biomembr. 2017, 1859:619-38. [DOI]
Y. M. Klein, A. Lanzilotto, A. Prescimone, K. W. Krämer, S. Decurtins, S. Liu, E. C. Constable, C. E. Housecroft “Coordination behaviour of 1-(3,2':6',3''-terpyridin-4'-yl)ferrocene: structure and magnetic and electrochemical properties of a tetracopper dimetallomacrocycle“ Polyhedron 2017, 129:71-76. [DOI]
I. Craciun, G. Gunkel-Grabole, A. Belluati, C. G. Palivan, W. Meier “Expanding the potential of MRI contrast agents through multifunctional polymeric nanocarriers“ Nanomedicine 2017, 12:811-17. [DOI]
D. Alsteens, H. E. Gaub, R. Newton, M. Pfreundschuh, C. Gerber, D. J. Müller “Atomic force microscopy-based characterization and design of biointerfaces“ Nat. Rev. Mater. 2017, 2(5):17008. [DOI]
B. M. Gaub, D. J. Müller “Mechanical Stimulation of Piezo1 Receptors Depends on Extracellular Matrix Proteins and Directionality of Force“ Nano Lett. 2017, 17(3):2064-72
M. Skaisgirski, X. Guo, O. Wenger “Electron Accumulation on Naphthalene Diimide Photosensitized by [Ru(2,2′-Bipyridine)3]2+“ Inorg. Chem. 2017, 56:2432-39. [DOI]
S. MatileC. G. Palivan “Strain-Promoted Thiol-Mediated Cellular Uptake of Giant Substrates: Liposomes and Polymersomes“ Angew. Chem. Int. Ed. 2017, 56:2947-50. [DOI]
D. J. Müller, H. E. Gaub “Membrane proteins scrambling through a folding landscape“ Science 2017. [DOI]
N. Zhang, V. Milleret, G. Thompson-Steckel, N. Huang, J. Vörös, B. R. Simona, M. Ehrbar “Soft Hydrogels Featuring In-Depth Surface Density Gradients for the Simple Establishment of 3D Tissue Models for Screening Applications“ SLAS Discov. 2017, 22:635-44. [DOI]
S. Lörcher, W. Meier “Cosolvent fractionation of PMOXA-b-PDMS-b-PMOXA: Bulk separation of triblocks from multiblocks“ Eur. Polym. J. 2017. [DOI]
E. V. Konishcheva, U. E. Zhumaev, W. P. Meier “PEO-b-PCL-b-PMOXA Triblock Copolymers: From Synthesis to Microscale Polymersomes with Asymmetric Membrane“ Macromolecules 2017. [DOI]
P. Baumann, M. Spulber, O. Fischer, A. Car, W. Meier “Investigation of Horseradish Peroxidase Kinetics in an “Organelle-Like” Environment“ Small 2017, 13:1603943. [DOI] [More Information]
P. Morelli, S. Matile “Sidechain Engineering in Cell-Penetrating Poly(disulfide)s“ Helv. Chim. Acta 2017, 100. [DOI]
P. Mohammadi, N. Beerenwinkel, Y. Benenson “Automated Design of Synthetic Cell Classifier Circuits Using a Two-Step Optimization Strategy“ 2017. [DOI]
D. Gregorowius, N. Biller‐Andorno, A. Deplazes Zemp “The role of scientific self‐regulation for the control of genome editing in the human germline“ EMBO Rep. 2017, 18:355-58. [DOI]
M. Macchione, N. Chuard, N. Sakai, S. Matile “Planarizable Push–Pull Probes: Overtwisted Flipper Mechanophores“ ChemPlusChem 2017. [DOI]
R. Wehlauch, S. M. Grendelmeier, H. Miyatake-Ondozabal, A. H. Sandtorv, M. Scherer, K. Gademann “Investigating Biogenetic Hypotheses of the Securinega Alkaloids: Enantioselective Total Syntheses of Secu’amamine E/ent-Virosine A and Bubbialine“ Org. Lett. 2017, 19:548-51. [DOI]
Q. Zhang, L. Catti, V. R. I. Kaila, K. Tiefenbacher “To catalyze or not to catalyze: elucidation of the subtle differences between the hexameric capsules of pyrogallolarene and resorcinarene“ Chem. Sci. 2017. [DOI]
M. Bharadwaj, N. Strohmeyer, G. P. Colo, J. Helenius, N. Beerenwinkel, H. B. Schiller, R. Fässler, D. J. Müller “αV-class integrins exert dual roles on α5β1 integrins to strengthen adhesion to fibronectin“ Nat. Commun. 2017. [DOI]
Q. Verolet, M. Dal Molin, A. Colom, A. Roux, L. Guénée, N. Sakai, S. Matile “Twisted Push‐Pull Probes with Turn‐On Sulfide Donors“ Helv. Chim. Acta 2017, 100:e1600328. [DOI]
A. Pannwitz, A. Prescimone, O. Wenger “Ruthenium(II)–Pyridylimidazole Complexes as Photoreductants and PCET Reagents“ Eur. J. Inorg. Chem. 2017, 2017:609-15. [DOI]
L. A. Büldt, X. Guo, R. Vogel, A. Prescimone, O. Wenger “A Tris (diisocyanide) chromium (0) Complex Is a Luminescent Analog of Fe (2, 2′-Bipyridine) 32+“ J. Am. Chem. Soc. 2017, 139:985-92. [DOI]
M. Muller, D. Auslander, A. Spinnler, J. Sikorski, M. Folcher, M. Fussenegger “Designed cell consortia as fragrance-programmable analog-to-digital converters“ 2017, 13:309-16. [DOI]
R. L. Stoop, K. Thodkar, M. Sessolo, H. J. Bolink, C. Schönenberger, M. Calame “Charge Noise in Organic Electrochemical Transistors“ Phys. Rev. Appl. 2017, 7:014009. [DOI]
S. Schuerle, I. A. Vizcarra, J. Moeller, M. S. Sakar, B. Özkale, A. M. Lindo, F. Mushtaq, I. Schoen, S. Pané, V. Vogel, B. J. Nelson “Robotically controlled microprey to resolve initial attack modes preceding phagocytosis“ Sci. Rob. 2017, 2:eaah6094. [DOI]
M. Jeschek, M. O. Bahls, V. Schneider, P. Marlière, T. R. WardS. Panke “Biotin-independent Strains of Escherichia coli for Enhanced Streptavidin Production“ Metab. Eng. 2017, 40:33-40. [DOI]
M. Kuss-Petermann, O. Wenger “Pump-Pump-Probe Spectroscopy of a Molecular Triad Monitoring Detrimental Processes for Photoinduced Charge Accumulation“ Helv. Chim. Acta 2017, 100:e1600283. [DOI]
J. Schreiber, M. Arter, N. Lapique, B. Haefliger, Y. Benenson “Model‐guided combinatorial optimization of complex synthetic gene networks“ Mol. Syst. Biol. 2016, 12:899. [DOI]
K. M. Carroll, H. Wolf, A. Knoll, J. E. Curtis, Y. Zhang, S. R. Marder, E. Riedo, U. Dürig “Understanding How Charged Nanoparticles Electrostatically Assemble and Distribute in 1-D“ Langmuir 2016, 32:13600-10. [DOI]
S. Soleimanpour, A. Colom, E. Derivery, M. Gonzalez-Gaitan, A. Roux, N. Sakai, S. Matile “Headgroup engineering in mechanosensitive membrane probes“ Chem. Commun. 2016, 52:14450-53. [DOI]
M. Xie, H. Ye, H. Wang, G. Charpin-El Hamri, C. Lormeau, P. Saxena, J. StellingM. Fussenegger “β-cell–mimetic designer cells provide closed-loop glycemic control“ Science 2016, 354:1296-1301. [DOI]
D. Gregorowius, A. Deplazes-Zemp “Societal impact of synthetic biology: responsible research and innovation (RRI)“ Essays Biochem. 2016, 60:371-79. [DOI]
C. Wang, F. N. Miros, J. Mareda, N. Sakai, S. Matile “Asymmetric Anion–π Catalysis on Perylenediimides“ Angew. Chem. Int. Ed. 2016, 55:14422-26. [DOI]
Y. M. Klein, A. Prescimone, E. C. Constable, C. E. Housecroft “Coordination Behaviour of 1-(4, 2′: 6′, 4 ″-terpyridin-4′-yl) ferrocene and 1-(3, 2′: 6′, 3 ″-terpyridin-4′-yl) ferrocene: Predictable and Unpredictable Assembly Algorithms“ Aust. J. Chem. 2016, 70:468. [DOI]
L. O. Herrmann, A. Olziersky, C. Gruber, G. Puebla-Hellmann, U. Drechsler, T. von Arx, K. Venkatesan, L. Novotny, E. Lörtscher “Fabrication of NEMS actuated plasmonic antenna platform for the study of optical forces and field enhancements in hot-spots“ Asia Communications and Photonics Conference 2016 2016:AS4B.2. [DOI]
T. Serdiuk, D. Balasubramaniam, J. Sugihara, S. A. Mari, H. R. Kaback, D. J. Müller “YidC assists the stepwise and stochastic folding of membrane proteins“ Nat. Chem. Biol. 2016, 12:911-17. [DOI]
D. Alsteens, R. Newton, R. Schubert, D. Martinez-Martin, M. Delguste, B. RoskaD. J. Müller “Nanomechanical mapping of first binding steps of a virus to animal cells“ Nat. Nanotechnol. 2016:DOI:10.1038/nnano.2016.228. [DOI]
R. Gnügge, L. Dharmarajan, M. Lang, J. Stelling “An Orthogonal Permease–Inducer–Repressor Feedback Loop Shows Bistability“ ACS Synth. Biol. 2016, 5:1098-107. [DOI]
J. Liu, V. Postupalenko, S. Lörcher, D. Wu, M. Chami, W. Meier, C. G. Palivan “DNA-Mediated Self-Organization of Polymeric Nanocompartments Leads to Interconnected Artificial Organelles“ Nano Lett. 2016:DOI: 10.1021/acs.nanolett.6b03430. [DOI] [More Information]
D. Vasquez, T. Einfalt, W. Meier, C. G. Palivan “Asymmetric Triblock Copolymer Nanocarriers for Controlled Localization and pH-Sensitive Release of Proteins“ Langmuir 2016, 32:10235-43. [DOI]
R. Kojima, D. Aubel, M. Fussenegger “Toward a world of theranostic medication: Programming biological sentinel systems for therapeutic intervention“ Adv. Drug Delivery Rev. 2016, 105:66-76. [DOI]
Z. Liu, V. Lebrun, T. Kitanosono, H. Mallin, V. Köhler, D. Häussinger, D. Hilvert, S. Kobayashi, T. R. Ward “Upregulation of an Artificial Zymogen by Proteolysis“ Angew. Chem. Int. Ed. 2016, 55:11587-90. [DOI]
N. Chuard, K. Fujisawa, P. Morelli, J. Saarbach, N. Winssinger, P. Metrangolo, G. Resnati, N. Sakai, S. Matile “Activation of Cell-Penetrating Peptides with Ionpair−π Interactions and Fluorophiles“ J. Am. Chem. Soc. 2016, 138:11264-71. [DOI]
L. A. Büldt, X. Guo, A. Prescimone, O. Wenger “A Molybdenum (0) Isocyanide Analogue of Ru (2, 2′‐Bipyridine) 32+: A Strong Reductant for Photoredox Catalysis“ Angew. Chem. Int. Ed. 2016, 55:11247-50. [DOI]
M. Jeschek, R. Reuter, T. Heinisch, C. Trindler, J. Klehr, S. PankeT. R. Ward “Directed evolution of artificial metalloenzymes for in vivo metathesis“ Nature 2016, doi:10.1038/nature19114. [DOI] [More Information]
M. Pogson, C. Parola, W. J. Kelton, P. Heuberger, S. Reddy “Immunogenomic engineering of a plug-and-(dis)play hybridoma platform“ Nat. Commun. 2016, 7:12535. [DOI]
M. Hestericová, M. R. Correro, M. Lenz, P. F. Corvini, P. Shahgaldian, T. R. Ward “Immobilization of an artificial imine reductase within silica nanoparticles improves its performance“ Chem. Commun. 2016, 52:9462-65. [DOI]
S. J. Sigg, F. Santini, A. Najer, P. U. Richard, W. P. Meier, C. G. Palivan “Nanoparticle-based highly sensitive MRI contrast agents with enhanced relaxivity in reductive milieu“ Chem. Commun. 2016, 52:9937-40. [DOI]
Z. P. Guven, B. Ustbas, K. M. Harkness, H. Coskun, C. P. Joshi, T. M. D. Besong, F. Stellacci, O. M. Bakr, O. Akbulut “Synthesis and characterization of mixed ligand chiral nanoclusters“ Dalton Trans. 2016, 45:11297-300. [DOI]
S. Benz, M. Macchione, Q. Verolet, J. Mareda, N. Sakai, S. Matile “Anion Transport with Chalcogen Bonds“ J. Am. Chem. Soc. 2016, 138:9093-96. [DOI]
S. G. Keller, A. Pannwitz, F. Schwizer, J. Klehr, O. WengerT. R. Ward “Light-driven electron injection from a biotinylated triarylamine donor to [Ru (diimine) 3] 2+-labeled streptavidin“ Org. Biomol. Chem. 2016, 14:7197-201. [DOI]
D. Harder, S. Hirschi, Z. Ucurum, R. Goers, W. Meier, D. J. MüllerD. Fotiadis “Engineering a Chemical Switch into the Light-driven Proton Pump Proteorhodopsin by Cysteine Mutagenesis and Thiol Modification“ Angew. Chem. Int. Ed.  2016, 55:8846. [DOI]
A. Najer, D. Wu, M. G. Nussbaumer, G. Schwertz, A. Schwab, M. C. Witschel, A. Schäfer, F. Diederich, M. Rottmann, C. G. Palivan, H. Beck, W. Meier “An amphiphilic graft copolymer-based nanoparticle platform for reduction-responsive anticancer and antimalarial drug delivery“ Nanoscale 2016:DOI: 10.1039/C6NR04290B. [DOI]
Y. OkamotoT. R. WardO. Wenger “From Photodriven Charge Accumulation to Fueling Enzyme Cascades in Molecular Factories“ Chimia 2016, 6:395. [DOI]
S. HirschiM. Stauffer, D. Harder, D. J. Müller, W. Meier, D. Fotiadis “Engineering and Assembly of Protein Modules into Functional Molecular Systems“ Chimia 2016, 6:398. [DOI]
Y. Cotelle, N. Chuard, S. Lascano, V. Lebrun, R. Wehlauch, N. Bohni, S. Lörcher, V. Postupalenko, S. Reddy, W. Meier, C. G. Palivan, K. Gademann, T. R. WardS. Matile “Interfacing Functional Systems“ Chimia 2016, 6:418. [DOI]
P. Rottmann, T. R. WardS. Panke “Compartmentalization – A Prerequisite for Maintaining and Changing an Identity“ Chimia 2016, 6:428
C. E. HousecroftC. G. Palivan, K. Gademann, W. Meier, M. Calame, V. Mikhalevich, X. Zhang, E. Piel, M. Szponarski, A. Wiesler, A. Lanzilotto, E. C. Constable, A. Fanget, R. Stoop “‘Active surfaces’ as Possible Functional Systems in Detection and Chemical (Bio) Reactivity“ Chimia 2016, 6:402. [DOI]
G. Puebla-Hellmann, M. MayorE. Lörtscher “Functional Nanopores: A Solid-state Concept for Artificial Reaction Compartments and Molecular Factories“ Chimia 2016, 6:432. [DOI]
W. Kelton, T. PeschS. MatileS. Reddy “Surveying the Delivery Methods of CRISPR/Cas9 for ex vivo Mammalian Cell Engineering“ Chimia 2016, 6:439. [DOI]
M. Garni, T. Einfalt, M. Lomora, A. Car, W. Meier, C. G. Palivan “Artificial Organelles: Reactions inside Protein–Polymer Supramolecular Assemblies“ Chimia 2016, 6:424. [DOI]
S. Vujica, C. Zelmer, R. Panatala, R. Y. H. Lim “Nucleocytoplasmic Transport: A Paradigm for Molecular Logistics in Artificial Systems“ Chimia 2016, 6:413. [DOI]
A. Deplazes Zemp “Artificial Cell Research as a Field that Connects Chemical, Biological and Philosophical Questions“ Chimia 2016, 6:443. [DOI]
R. Heidari, B. Elger, R. Stutzki “On the Brink of Shifting Paradigms, Molecular Systems Engineering Ethics Needs to Take a Proactive Approach“ Chimia 2016, 6:449. [DOI]
Y. Benenson “Complexity from Simple Building Blocks: Engineering Large-scale Information-processing Networks from Molecules“ Chimia 2016, 6:392. [DOI]
L. Liu, Y. Cotelle, A. Avestro, N. Sakai, S. Matile “Asymmetric Anion−π Catalysis of Iminium/Nitroaldol Cascades To Form Cyclohexane Rings with Five Stereogenic Centers Directly on π-Acidic Surfaces“ J. Am. Chem. Soc. 2016, 138:7876-79. [DOI]
T. M. Roberts, F. Rudolf, A. Meyer, R. Pellaux, E. Whitehead, S. Panke, M. Held “Identification and Characterisation of a pH-stable GFP“ Sci. Rep. 2016, 6:28166. [DOI]
A. Bolag, N. Sakai, S. Matile “Dipolar Photosystems: Engineering Oriented Push–Pull Components into Double- and Triple-Channel Surface Architectures“ Chem. Eur. J. 2016, 22:9006-14. [DOI]
M. JeschekS. PankeT. R. Ward “Chapter Twenty-Three-Periplasmic Screening for Artificial Metalloenzymes“ Methods Enzymol. 2016, 580:539-56. [DOI]
R. Boggavarapu, S. Hirschi, D. Harder, M. Meury, Z. Ucurum, M. J. Bergeron, D. Fotiadis “Purification of Human and Mammalian Membrane Proteins Expressed in Xenopus laevis Frog Oocytes for Structural Studies“ Heterologous Expression of Membrane Proteins 2016, 1432:223-42. [DOI]
J. Foolen, J. Shiu, M. Mitsi, Y. Zhang, C. S. Chen, V. Vogel “Full-Length Fibronectin Drives Fibroblast Accumulation at the Surface of Collagen Microtissues during Cell-Induced Tissue Morphogenesis“ PLoS One 2016, 11:e0160369. [DOI]
Y. M. Klein, A. Prescimone, M. B. Pitak, S. J. Coles, E. C. Constable, C. E. Housecroft “Constructing chiral MOFs by functionalizing 4, 2′: 6′, 4′′-terpyridine with long-chain alkoxy domains: rare examples of neb nets“ CrystEngComm 2016, 18:4704-07. [DOI]
L. Liu, S. Matile “Anion-π transaminase mimics“ Supramol. Chem. 2016:1-5. [DOI]
V. Mikhalevich, C. Zelmer, A. Car, C. G. Palivan, W. Meier “Bio-inspired Polymer Membranes“ Bio-inspired Polymers 2016, 22:221-58. [DOI]
E. A. Miłopolska, M. Kuss-Petermann, M. Neuburger, O. WengerT. R. Ward “N-Heterocyclic carbene ligands bearing a naphthoquinone appendage: Synthesis and coordination chemistry“ Polyhedron 2016, 103:261-66. [DOI]
P. Morelli, X. Martin-Benlloch, R. Tessier, J. Waser, N. Sakai, S. Matile “Ethynyl benziodoxolones: functional terminators for cell-penetrating poly(disulfide)s“ Polym. Chem. 2016, 7:3465-70. [DOI]
M. Pogson, W. Kelton, S. Reddy “Microscale Technologies for High-Throughput Analysis of Immune Cells“ Microscale Technologies for Cell Engineering 2016:219-30. [DOI]
Y. Cotelle, V. Lebrun, N. Sakai, T. R. WardS. Matile “Anion‑π Enzymes“ ACS Cent. Sci. 2016:DOI: 10.1021/acscentsci.6b00097. [DOI] [More Information]
M. Wipf, R. L. Stoop, G. Navarra, S. Rabbani, B. Ernst, K. Bedner, C. Schönenberger, M. Calame “Label-Free FimH Protein Interaction Analysis Using Silicon Nanoribbon BioFETs“ ACS Sens. 2016:DOI: 10.1021/acssensors.6b00089. [DOI]
T. Raschle, P. Rios Flores, C. Opitz, D. J. Müller, S. Hiller “Monitoring Backbone Hydrogen-Bond Formation in β-Barrel Membrane Protein Folding“ Angew. Chem. Int. Ed. 2016, 55:5952-55. [DOI]
R. L. Stoop, M. Wipf, S. Müller, K. Bedner, I. A. Wright, C. J. Martin, E. C. Constable, A. Fanget, C. Schönenberger, M. Calame “Implementing Silicon Nanoribbon Field-Effect Transistors as Arrays for Multiple Ion Detection“ Biosensors 2016, 6:21. [DOI]
Y. Sakiyama, A. Mazur, L. E. Kapinos, R. Y. H. Lim “Spatiotemporal dynamics of the nuclear pore complex transport barrier resolved by high-speed atomic force microscopy“ Nat. Nanotechnol. 2016:DOI: 10.1038/nnano.2016.62. [DOI]
N. Strelnikova, F. Herren, C. Schoenenberger, T. Pfohl “Formation of Actin Networks in Microfluidic Concentration Gradients“ Front. Mater. 2016, 3. [DOI]
Y. Okamoto, V. Köhler, C. E. Paul, F. Hollmann, T. R. Ward “Efficient In Situ Regeneration of NADH Mimics by an Artificial Metalloenzyme“ ACS Catal. 2016, 6:3553. [DOI]
M. Xie, V. Haellman, M. FusseneggerV. Hällman “Synthetic biology-application-oriented cell engineering.“ Curr. Opin. Biotechnol. 2016, 40:139. [DOI]
I. A. Dinu, J. T. Duskey, A. Car, C. G. Palivan, W. Meier “Engineered non-toxic cationic nanocarriers with photo-triggered slow-release properties“ Polym. Chem. 2016, 7:3451. [DOI]
Y. Okamoto, V. Köhler, T. R. Ward “An NAD(P)H-Dependent Artificial Transfer Hydrogenase for Multienzymatic Cascades“ J. Am. Chem. Soc. 2016, 138:5781. [DOI]
S. Lascano, K. Zhang, R. Wehlauch, K. Gademann, N. Sakaiab, S. Matile “The third orthogonal dynamic covalent bond“ Chem. Sci. 2016, 7:4720. [DOI]
P. Saxena, B. C. Heng, P. Bai, M. Folcher, H. Zulewski, M. Fussenegger “A programmable synthetic lineage-control network that differentiates human IPSCs into glucose-sensitive insulin-secreting beta-like cells“ Nat. Commun. 2016, 7:DOI:10.1038/ncomms11247. [DOI] [More Information]
P. Bai, H. Ye, M. Xie, P. Saxena, H. Zulewski, G. Charpin-El Hamri, V. Djonov, M. Fussenegger “A synthetic biology-based device prevents liver injury in mice“ J. Hepatol. 2016, 65:84. [DOI]
M. Akamatsu, S. Matile “Expanded Chiral Surfaces for Asymmetric Anion–π Catalysis“ Synlett 2016, 27:1041-46. [DOI]
Y. Zhao, Y. Cotelle, N. Sakai, S. Matile “Unorthodox Interactions at Work“ J. Am. Chem. Soc. 2016, 138:4270-77. [DOI]
L. Ge, S. Villinger, S. Mari, K. Giller, C. Griesinger, S. Becker, D. J. Müller, M. Zweckstetter “Molecular Plasticity of the Human Voltage-Dependent Anion Channel Embedded Into a Membrane“ Structure 2016, 24:585-94. [DOI]
A. Najer, S. Thamboo, C. G. Palivan, H. Beck, W. Meier “Giant Host Red Blood Cell Membrane Mimicking Polymersomes Bind Parasite Proteins and Malaria Parasites“ Chimia 2016, 4:288. [DOI]
H. Mallin, M. Hestericová, R. Reuter, T. R. Ward “Library design and screening protocol for artificial metalloenzymes based on the biotin-streptavidin technology“ Nat. Protoc. 2016, 11:835. [DOI]
M. JeschekD. GerngrossS. Panke “Rationally reduced libraries for combinatorial pathway optimization minimizing experimental effort“ Nat. Commun. 2016, 7:doi:10.1038/ncomms11163. [DOI]
M. Hilbert, A. Noga, D. Frey, V. Hamel, P. Guichard, S. H. Kraatz, M. Pfreundschuh, S. Hosner, I. Flückiger, R. Jaussi, M. M. Wieser, K. M. Thieltges, X. Deupi, D. J. Mülle, R. Kammerer, P. Gönczy, M. Hirono, M. O. Steinmetz, D. J. Müller “SAS-6 engineering reveals interdependence between cartwheel and microtubules in determining centriole architecture“ Nat. Cell Biol. 2016, 18:393. [DOI]
C. Albayrak, C. A. Jordi, C. Zechner, J. Lin, C. A. Bichsel, M. Khammash, S. Tay “Digital Quantification of Proteins and mRNA in Single Mammalian Cells“ Mol. Cell 2016, 61:791. [DOI]
S. J. Sigg, V. Postupalenko, J. T. Duskey, C. G. Palivan, W. Meier “Stimuli-Responsive Codelivery of Oligonucleotides and Drugs by Self-Assembled Peptide Nanoparticles“ Biomacromolecules 2016, 17:935-45. [DOI]
D. Auslaender, D. W. Fuchs, S. Huerlemann, M. Fussenegger “Engineering a ribozyme cleavage-induced split fluorescent aptamer complementation assay“ Nucleic Acids Res. 2016, 44:e94. [DOI]
X. Zhang, M. Lomora, T. Einfalt, W. Meier, N. Klein, D. Schneider, C. G. Palivan “Active surfaces engineered by immobilizing protein-polymer nanoreactors for selectively detecting sugar alcohols“ Biomaterials 2016, 89:79. [DOI]
Y. Cotelle, S. Benz, A. Avestro, T. R. Ward, N. Sakai, S. Matile “Anion-π Catalysis of Enolate Chemistry: Rigidified Leonard Turns as a General Motif to Run Reactions on Aromatic Surfaces“ Angew. Chem. Int. Ed. 2016, 55:4275. [DOI]
L. P. Zweifel, I. Shorubalko, R. Y. Lim “Helium Scanning Transmission Ion Microscopy and Electrical Characterization of Glass Nanocapillaries with Reproducible Tip Geometries“ ACS Nano 2016, 10:1918-25. [DOI]
F. N. Miros, Y. Zhao, G. Sargsyan, M. Pupier, C. Besnard, C. Beuchat, J. Mareda, N. Sakai, S. Matile “Enolate Stabilization by Anion–π Interactions: Deuterium Exchange in Malonate Dilactones on π-Acidic Surfaces“ Chem. Eur. J. 2016, 22:2648-57. [DOI]
M. A. Burkhardt, J. Waser, V. Milleret, I. Gerber, M. Y. Emmert, J. Foolen, S. P. Hoerstrup, F. Schlottig, V. Vogel “Synergistic interactions of blood-borne immune cells, fibroblasts and extracellular matrix drive repair in an in vitro peri-implant wound healing model“ Sci. Rep. 2016, 6:21071. [DOI]
B. Häfliger, L. Prochazka, B. Angelici, Y. Benenson “Precision multidimensional assay for high-throughput microRNA drug discovery“ Nat. Commun. 2016, 7:DOI: 10.1038/ncomms10709. [DOI] [More Information]
A. Tastanova, A. Schulz, A. Tolstrup, A. Puklowski, H. Kaufmann, M. Fussenegger “Overexpression of YY1 increases the protein production in mammalian cells“ J. Biotechnol. 2016, 219:72. [DOI]
G. Gunkel-Grabole, A. Car, V. V. Naik, L. Marot, G. Ferk, C. G. Palivan, W. Meier “PEG Brushes on Porous, PDMS-Coated Surfaces and Their Interaction with Carbon Dioxide“ Macromol. Chem. Phys. 2016, 217:966. [DOI]
A. Lanzilotto, L. A. Büldt, H. C. Schmidt, A. Prescimone, O. Wenger, E. C. Constable, C. E. Housecroft “Improved light absorbance does not lead to better DSC performance: studies on a ruthenium porphyrin–terpyridine conjugate“ RSC Adv. 2016, 6:15370. [DOI]
P. Saxena, G. Charpin-El Hamri, M. Folcher, H. Zulewski, M. Fussenegger “Synthetic gene network restoring endogenous pituitary-thyroid feedback control in experimental Graves' disease“ Proc. Natl. Acad. Sci. U.S.A. 2016, 113:1244. [DOI]
F. Schwarz, G. Kastlunger, F. Lissel, C. Egler-Lucas, S. N. Semenov, K. Venkatesan, H. Berke, R. Stadler, E. Lortscher “Field-induced conductance switching by charge-state alternation in organometallic single-molecule junctions“ Nat. Nanotechnol. 2016, 11:170-76. [DOI]
S. Benz, J. López‐Andarias, J. Mareda, N. Sakai, S. Matile “Catalysis with Chalcogen Bonds“ Angew. Chem. Int. Ed. 2016, 56:812-15. [DOI]
H. C. Schmidt, M. Spulber, M. Neuburger, C. G. Palivan, M. Meuwly, O. Wenger “Charge Transfer Pathways in Three Isomers of Naphthalene-Bridged Organic Mixed Valence Compounds“ J. Org. Chem. 2016, 81:595-602. [DOI]
Y. M. Klein, A. Prescimone, E. C. Constable, C. E. Housecroft “2-Dimensional networks assembled using 4′-functionalized 4,2′:6′,4″-terpyridines and Co(NCS)2“ Polyhedron 2016, 103:58. [DOI]
K. Yonehara1, M. Fiscella, A. Drinnenberg, F. Esposti, S. Trenholm, J. Krol, F. Franke, C. Gross Scherf, A. Kusnyerik, J. Müller, A. Szabo, J. Jüttner, F. Cordoba, A. P. Reddy, J. Németh, Z. Z. Nagy, F. Munier, A. Hierlemann, B. Roska “Congenital Nystagmus Gene FRMD7 Is Necessary for Establishing a Neuronal Circuit Asymmetry for Direction Selectivity“ Neuron 2016, 89:177. [DOI]
I. Avalos Vizcarra, V. Hosseini, P. Kollmannsberger, S. Meier, S. S. Weber, M. Arnoldini, M. Ackermann, V. Vogel “How type 1 fimbriae help Escherichia coli to evade extracellular antibiotics“ Sci. Rep. 2016, 6:DOI: 10.1038/srep18109. [DOI]
B. Geering, Z. Zokouri, S. Hurlemann, B. Gerrits, D. Auslander, A. Britschgi, M. P. Tschan, H. U. Simon, M. Fussenegger “Identification of Novel Death-Associated Protein Kinase 2 Interaction Partners by Proteomic Screening Coupled with Bimolecular Fluorescence Complementation“ Mol. Cell. Biol. 2016, 36:132-43. [DOI]
T. Heinisch, M. Pellizzoni, M. Dürrenberger, C. E. Tinberg, V. Köhler, J. Klehr, D. Häussinger, D. Baker, T. R. Ward “Improving the Catalytic Performance of an Artificial Metalloenzyme by Computational Design“ J. Am. Chem. Soc. 2016, 137:10414. [DOI]
A. Chatterjee, H. Mallin, J. Klehr, J. Vallapurackal, A. D. Finke, L. Vera, M. Marsh, T. R. Ward “An enantioselective artificial Suzukiase based on the biotin–streptavidin technology “ Chem. Sci. 2016, 7:673. [DOI]
Q. Verolet, A. Rosspeintner, S. Soleimanpour, N. Sakai, E. Vauthey, S. Matile “Turn-On Sulfide π Donors: An Ultrafast Push for Twisted Mechanophores“ J. Am. Chem. Soc. 2015, 137:15644-47. [DOI]
L. Lina Schukur, B. Geering, G. Charpin-El Hamri, M. Fussenegger “Implantable synthetic cytokine converter cells with AND-gate logic treat experimental psoriasis“ Sci. Transl. Med. 2015, 7:DOI: 10.1126/scitranslmed.aac4964. [DOI]
G. Gasparini, S. Matile “Protein delivery with cell-penetrating poly(disulfide)s“ Chem. Commun. 2015, 51:17160-62. [DOI]
R. L. Stoop, M. Wipf, S. Mueller, K. Bedner, I. A. Wright, C. J. Martin, E. C. Constable, W. Fu, A. Tarasov, M. Calame, C. Schönenberger “Competing surface reactions limiting the performance of ion-sensitivefield-effect transistors“ Sens. Actuators, B 2015, 220:500. [DOI]
B. C. Heng, D. Aubel, M. Fussenegger “Prosthetic gene networks as an alternative to standard pharmacotherapies for metabolic disorders“ Curr. Opin. Biotechnol. 2015, 35:37-45. [DOI]
B. Source, C. Escobedo, Y. Toyoda, M. P. Stewart, C. J. Cattin, R. Newton, I. Banerjee, A. Stettler, B. Roska, S. Eaton, A. A. Hyman, A. Hierlemann, D. J. Müller “Mitotic cells contract actomyosin cortex and generate pressure to round against or escape epithelial confinement“ Nat. Commun. 2015, 6:8872. [DOI] [More Information]
P. Muller, M. Kreuzaler, T. Khan, D. S. Thommen, K. Martin, K. Glatz, S. Savic, N. Harbeck, U. Nitz, O. Gluz, M. von Bergwelt-Baildon, H. Kreipe, S. Reddy, M. Christgen, A. Zippelius “Trastuzumab emtansine (T-DM1) renders HER2+ breast cancer highly susceptible to CTLA-4/PD-1 blockade“ Sci. Transl. Med. 2015, 7:315ra188. [DOI]
C. G. Palivan, R. Goers, A. Najer, X. Zhang, A. Car, W. Meier “Bioinspired polymer vesicles and membranes for biological and medical applications“ Chem. Soc. Rev. 2015, 45:377. [DOI]
M. Pfreundschuh, D. Alsteens, R. Wieneke, C. Zhang, S. R. Coughlin, R. Tampé, B. K. Kobilka, D. J. Müller “Identifying and quantifying two ligand-binding sites while imaging native human membrane receptors by AFM“ Nat. Commun. 2015, 6:8857. [DOI]
A. Najer, S. Thamboo, J. T. Duskey, W. Meier, C. G. Palivan “Analysis of Molecular Parameters Determining the Antimalarial Activity of Polymer-Based Nanomimics“ Macromol. Rapid Commun. 2015, 36:1923. [DOI]
P. Rajendran, S. Kaufmann, J. Vörös, M. Zenobi-Wong, L. Demkó “Femtomolar oligonucleotide detection by a one-step gold nanoparticle-based assay“ Colloids Surf., B 2015, 135:193. [DOI]
R. Reuter, T. R. Ward “Profluorescent substrates for the screening of olefin metathesis catalysts“ Beilstein J. Org. Chem. 2015, 11:1886-92. [DOI]
T. Einfalt, R. Goers, I. A. Dinu, A. Najer, M. Spulber, O. Onaca-Fischer, C. G. Palivan “Stimuli-triggered activity of nano reactors by biomimetic engineering polymer membranes“ Nano Lett. 2015, 15:7596. [DOI] [More Information]
M. Fiscella, F. Franke, K. Farrow, J. Muller, B. Roska, R. A. da Silveira, A. Hierlemann “Visual coding with a population of direction-selective neurons“ J. Neurophysiol. 2015, 114:2485-99. [DOI]
L. Schukur, B. Geering, M. Fussenegger “Human whole-blood culture system for ex vivo characterization of designer-cell function“ Biotechnol. Bioeng. 2015, 113:588. [DOI]
Y. Zhao, Y. Cotelle, A. J. Avestro, N. Sakai, S. Matile “Asymmetric Anion-π Catalysis: Enamine Addition to Nitroolefins on pi-Acidic Surfaces“ J. Am. Chem. Soc. 2015, 137:11582-85. [DOI]
J. Thoma, B. M. Burmann, S. Hiller, D. J. Müller “Impact of holdase chaperones Skp and SurA on the folding of β-barrel outer-membrane proteins“ Nat. Struct. Mol. Biol. 2015, 22:795. [DOI] [More Information]
K. Fujisawa, M. Humbert-Droz, R. Letrun, E. Vauthey, T. A. Wesolowski, N. Sakai, S. Matile “Ion Pair-π Interactions“ J. Am. Chem. Soc. 2015, 137:11047-56. [DOI]
V. Haellman, M. FusseneggerV. Hällman “Synthetic Biology-Toward Therapeutic Solutions“ J. Mol. Biol. 2015, 428:945. [DOI]
A. Adamantidis, S. Arber, J. S. Bains, E. Bamberg, A. Bonci, G. Buzsaki, J. A. Cardin, R. M. Costa, Y. Dan, Y. Goda, A. M. Graybiel, M. Hausser, P. Hegemann, J. R. Huguenard, T. R. Insel, P. H. Janak, D. Johnston, S. A. Josselyn, C. Koch, A. C. Kreitzer, C. Luscher, R. C. Malenka, G. Miesenbock, G. Nagel, B. Roska, M. J. Schnitzer, K. V. Shenoy, I. Soltesz, S. M. Sternson, R. Y. Tsien, G. G. Turrigiano, K. M. Tye, R. I. Wilson “Optogenetics: 10 years after ChR2 in neurons--views from the community“ Nat. Neurosci. 2015, 18:1202-12. [DOI]
D. Kalbermatter, J. Jeckelmann, P. Chiu, Z. Ucurum, T. Walz, D. Fotiadis “2D and 3D crystallization of the wild-type IIC domain of the glucose PTS transporter from Escherichia coli“ J. Struct. Biol. 2015, 191:376-80. [DOI]
M. Lomora, I. A. Dinu, F. Itel, S. Rigo, M. Spulber, C. G. Palivan “Does Membrane Thickness Affect the Transport of Selective Ions Mediated by Ionophores in Synthetic Membranes?“ Macromol. Rapid Commun. 2015, 36:1929. [DOI]
K. D. Zhang, N. Sakai, S. Matile “Colorful surface architectures with three different types of dynamic covalent bonds: integration of anthocyanins, tritylium ions and flavins“ Org. Biomol. Chem. 2015, 13:8687-94. [DOI]
E. Mulvihill, K. van Pee, S. A. Mari, D. J. Müller, O. Yildiz “Directly Observing the Lipid-Dependent Self-Assembly and Pore-Forming Mechanism of the Cytolytic Toxin Listeriolysin O“ Nano Lett. 2015, 15:6965. [DOI]
T. O. Ihalainen, L. Aires, F. A. Herzog, R. Schwartlander, J. Moeller, V. Vogel “Differential basal-to-apical accessibility of lamin A/C epitopes in the nuclear lamina regulated by changes in cytoskeletal tension“ Nat. Mater. 2015, 14:1252. [DOI]
J. Liu, V. Postupalenko, J. T. Duskey, C. G. Palivan, W. Meier, V. Mikhalevich “pH-Triggered Reversible Multiple Protein-Polymer Conjugation Based on Molecular Recognition“ J. Chem. Phys. B. 2015, 119:12066. [DOI]
H. Wang, H. Ye, M. Xie, M. Daoud El-Baba, M. Fussenegger “Cosmetics-triggered percutaneous remote control of transgene expression in mice“ Nucleic Acids Res. 2015, 43:e91. [DOI]
K. E. Kubow, R. Vukmirovic, L. Zhe, E. Klotzsch, M. L. Smith, D. Gourdon, S. Luna, V. Vogel “Mechanical forces regulate the interactions of fibronectin and collagen I in extracellular matrix“ Nat. Commun. 2015, 6:8026. [DOI]
D. Sun, T. Flock, X. Deupi, S. Maeda, M. Matkovic, S. Mendieta, D. Mayer, R. J. Dawson, G. Schertler, M. M. Babu, D. B. Veprintsev “Probing Gαi1 protein activation at single–amino acid resolution“ Nat. Struct. Mol. Biol. 2015, 22:686. [DOI]
R. Boggavarapu, J. Jeckelmann, D. Harder, Z. Ucurum, D. Fotiadis “Role of electrostatic interactions for ligand recognition and specificity of peptide transporters“ BMC Biol. 2015, 13:DOI: 10.1186/s12915-015-0167-8. [DOI] [More Information]
C. J. Cattin, M. Düggelin, M. Martinez-Martin, D. C. Gerber, D. J. Müller, M. P. Stewart “Mechanical control of mitotic progression in single animal cells “ Proc. Natl. Acad. Sci. U.S.A. 2015, 112:11258. [DOI]
C. M. Gruber, L. O. Herrmann, A. Olziersky, G. F. Puebla-Hellman, U. Drechsler, T. von Arx, M. Koch, Z. J. Lapin, K. Venkatesan, L. Novotna, E. Lortscher, E. Lörtscher “Fabrication of bow-tie antennas with mechanically tunable gap sizes below 5 nm for single-molecule emission and Raman scattering“ IEEE Nano 2015:20. [DOI]
G. Puebla-Hellman, M. Mayor, E. Lortscher, E. Lörtscher “Ultraflat nanopores for wafer-scale molecular-electronic applications“ IEEE Nano 2015:1197. [DOI]
K. D. Zhang, S. Matile “Complex Functional Systems with Three Different Types of Dynamic Covalent Bonds“ Angew. Chem. Int. Ed. 2015, 54:8980-83. [DOI]
D. Alsteens, M. Pfreundschuh, C. Zhang, P. M. Spoerri, S. R. Coughlin, B. K. Kobilka, D. J. Müller “Imaging G protein–coupled receptors while quantifying their ligand-binding free-energy landscape“ Nat. Methods 2015, 12:845. [DOI] [Author File on Daniel J. Müller]
Y. M. Klein, A. Prescimone, E. C. Constable, C. E. Housecroft “Manipulating connecting nodes through remote alkoxy chain variation in coordination networks with 4′-alkoxy-4,2′:6′,4′′-terpyridine linkers“ CrystEngComm 2015, 17:6483. [DOI]
G. Gasparini, E. K. Bang, J. Montenegro, S. Matile “Cellular uptake: lessons from supramolecular organic chemistry“ Chem. Commun. 2015, 51:10389-402. [DOI]
A. Wertz, S. Trenholm, K. Yonehara, D. Hillier, Z. Raics, M. Leinweber, G. Szalay, A. Ghanem, G. Keller, B. Rózsa, K. K. Conzelmann, B. Roska “Single-cell–initiated monosynaptic tracing reveals layer-specific cortical network modules“ Science 2015, 349:70. [DOI] [More Information]
T. Heinisch, T. R. Ward “Latest Developments in Metalloenzyme Design and Repurposing“ Eur. J. Inorg. Chem. 2015, 2015:3406-18. [DOI]
E. Kaufmann, H. Hattori, H. Miyatake-Ondozabal, K. Gademann “Total Synthesis of the Glycosylated Macrolide Antibiotic Fidaxomicin“ Org. Lett. 2015, 17:3514. [DOI]
D. Fotiadis, A. Engel “Two‐Dimensional Crystallisation of Membrane Proteins and Structural Assessment“ eLS 2015:1-10. [DOI]
G. Gasparini, G. Sargsyan, E. Bang, N. Sakai, S. Matile “Ring Tension Applied to Thiol‐Mediated Cellular Uptake“ Angew. Chem. Int. Ed. 2015, 54:7328-31. [DOI]
R. Kojima, D. Aubel, M. Fussenegger “Novel theranostic agents for next-generation personalized medicine: small molecules, nanoparticles, and engineered mammalian cells“ Curr. Opin. Chem. Biol. 2015, 28:29. [DOI]
S. M. Früh, I. Schoen, J. Ries, V. Vogel “Molecular architecture of native fibronectin fibrils“ Nat. Commun. 2015, 6:7275. [DOI]
I. Krol, C. P. Alvarez, M. Fiscella, A. Hierlemann, B. Roska, W. Filipowicz “A network comprising short and long noncoding RNAs and RNA helicase controls mouse retina architecture“ Nat. Commun. 2015, 6:7305. [DOI]
O. Yushchenko, G. Licari, S. Mosquera-Vazquez, N. Sakai, S. Matile, E. Vauthey “Ultrafast Intersystem-Crossing Dynamics and Breakdown of the Kasha-Vavilov's Rule of Naphthalenediimides“ J. Phys. Chem. Lett. 2015, 6:2096-100. [DOI]
S. Y. Brauchli, F. J. Malzner, E. C. Constable, C. E. Housecroft “Copper(I)-based dye-sensitized solar cells with sterically demanding anchoring ligands: bigger is not always better“ RSC Adv. 2015, 5:48516. [DOI]
T. Einfalt, G. Gunkel-Grabole, M. Spulber, A. Najer, C. G. Palivan “Supramolecular Architectures: Supramolecular Architectures from Self-Assembled Copolymers“ CRC Concise Encyclopedia of Nanotechnology 2015:1055-72. [DOI]
M. Plodinec, R. Y. Lim “Nanomechanical Characterization of Living Mammary Tissues by Atomic Force Microscopy“ Mammary Stem Cells 2015, 1293:231-46. [DOI]
Q. Verolet, S. Soleimanpour, K. Fujisawa, M. Dal Molin, N. Sakai, S. Matile “Design and Synthesis of Mixed Oligomers with Thiophenes, Dithienothiophene S, S‐Dioxides, Thieno [3, 4] pyrazines and 2, 1, 3‐Benzothiadiazoles: Flipper Screening for Mechanosensitive Systems“ ChemistryOpen 2015, 4:264-67. [DOI]
M. Yu, N. Strohmeyer, J. Wang, D. J. Müller, J. Helenius “Increasing throughput of AFM-based single cell adhesion measurements through multisubstrate surfaces“ Beilstein J. Nanotechnol. 2015, 6:157-66. [DOI]
O. Yushchenko, D. Villamaina, N. Sakai, S. Matile, E. Vauthey “Comparison of Charge-Transfer Dynamics of Naphthalenediimide Triads in Solution and π-Stack Architectures on Solid Surfaces“ J. Phys. Chem. B 2015, 119:14999-15008. [DOI]
Y. Zhao, S. Benz, N. Sakai, S. Matile “Selective acceleration of disfavored enolate addition reactions by anion–π interactions“ Chem. Sci. 2015, 6:6219-23. [DOI]
G. Tsiavaliaris, F. Itel, K. Hedfalk, S. Al-Samir, W. P. Meier, G. Gros, V. Endeward “Low CO2 permeability of cholesterol-containing liposomes detected by stopped-flow fluorescence spectroscopy“ FASEB J. 2015, 29:1780. [DOI]
J. Zhao, A. Kajetanowicz, T. R. Ward “Carbonic anhydrase II as host protein for the creation of a biocompatible artificial metathesase“ Org. Biomol. Chem. 2015, 13:5652-55. [DOI]
F. Itel, A. Najer, C. G. Palivan, W. Meier “Dynamics of Membrane Proteins within Synthetic Polymer Membranes with Large Hydrophobic Mismatch“ Nano Lett. 2015, 15:3871. [DOI] [More Information] [Artikel in ChimieXtra]
M. Xie, M. Fussenegger “Mammalian designer cells: Engineering principles and biomedical applications.“ Biotechnol. J. 2015, 10:1005. [DOI]
T. Frank, S. Tay “Automated co-culture system for spatiotemporal analysis of cell-to-cell communication“ Lab Chip 2015, 15:2192-200. [DOI]
R. Petrosyan, C. A. Bippes, S. Walheim, D. Harder, D. Fotiadis, T. Schimmel, D. Alsteens, D. J. Müller “Single-Molecule Force Spectroscopy of Membrane Proteins from Membranes Freely Spanning Across Nanoscopic Pores“ Nano Lett. 2015, 15:3624. [DOI]
T. Kim, M. Folcher, M. Doaud-El Baba, M. Fussenegger “A Synthetic Erectile Optogenetic Stimulator Enabling Blue-Light-Inducible Penile Erection“ Angew. Chem. Int. Ed. 2015, 54:5933. [DOI] [More Information]
E. Koren, E. Lörtscher, C. Rawlings, A. W. Knoll, U. Dürig “Surface science. Adhesion and friction in mesoscopic graphite contacts“ Science 2015, 348:679-83. [DOI]
N. Fujieda, J. Schätti, E. Stuttfeld, K. Ohkubo, T. Maier, S. Fukuzumi, T. R. Ward “Enzyme repurposing of a hydrolase as an emergent peroxidase upon metal binding“ Chem. Sci. 2015, 6:4060. [DOI]
J. Krol, B. Roska “Rods Feed Cones to Keep them Alive“ Cell 2015, 161:706-08. [DOI]
T. Kim, M. Folcher, G. Charpin-El Hamri, M. Fussenegger “A synthetic cGMP-sensitive gene switch providing Viagra((R))-controlled gene expression in mammalian cells and mice“ Metab. Eng. 2015, 29:169-79. [DOI]
J. Lopez-Andarias, A. Bolag, C. Nancoz, E. Vauthey, C. Atienza, N. Sakai, N. Martin, S. Matile “Electron-deficient fullerenes in triple-channel photosystems“ Chem. Commun. 2015, 51:7543-45. [DOI]
N. Hostettler, I. A. Wright, B. Bozic-Weber, E. C. Constable, C. E. Housecroft “Dye-sensitized solar cells with hole-stabilizing surfaces: “inorganic” versus “organic” strategies“ RSC Adv. 2015, 5:37906. [DOI]
A. Deplazes Zemp, D. Gregorowius, N. Biller-Andorno “Different Understandings of Life as an Opportunity to Enrich the Debate About Synthetic Biology“ Springer, Nanoethics 2015, 9:179. [DOI]
A. Hochstetter, E. Stellamanns, S. Deshpande, S. Uppaluri, M. Engstler, T. Pfohl “Microfluidics-based single cell analysis reveals drug-dependent motility changes in trypanosomes“ Lab Chip 2015, 15:1961-68. [DOI]
F. M. Heydenreich, Z. Vuckovic, M. Matkovic, D. B. Veprintsev “Stabilization of G protein-coupled receptors by point mutations“ Front. Pharmacol. 2015, 6:82. [DOI] [More Information]
G. Emilsson, R. L. Schoch, L. Feuz, F. Hook, R. Y. Lim, A. B. Dahlin “Strongly stretched protein resistant poly(ethylene glycol) brushes prepared by grafting-to“ ACS Appl. Mater. Interfaces 2015, 7:7505-15. [DOI]
M. Dal Molin, Q. Verolet, S. Soleimanpour, S. Matile “Mechanosensitive membrane probes“ Chem. Eur. J. 2015, 21:6012-21. [DOI]
Y. Zhao, G. Huang, C. Besnard, J. Mareda, N. Sakai, S. Matile “Big, Strong, Neutral, Twisted, and Chiral π Acids“ Chem. Eur. J. 2015, 21:6202-07. [DOI]
J. Kowal, D. Wu, V. MikhalevichC. G. Palivan, W. Meier “Hybrid Polymer–Lipid Films as Platforms for Directed Membrane Protein Insertion“ Langmuir 2015, 31:4868. [DOI]
T. Serdiuk, J. Sugihara, S. Mari, H. . Kaback, D. J. Müller “Observing a Lipid-Dependent Alteration in Single Lactose Permeases“ Structure 2015, 23:754-61. [DOI]
D. Milić, D. B. Veprintsev “Large-scale production and protein engineering of G protein-coupled receptors for structural studies“ Front. Pharmacol. 2015, 6:66. [DOI] [More Information]
M. Lomora, M. Garni, F. Itel, P. Tanner, M. Spulber, C. G. Palivan “Polymersomes with engineered ion selective permeability as stimuli-responsive nanocompartments with preserved architecture“ Biomaterials 2015, 53:406. [DOI]
M. Mehling, T. Frank, C. Albayrak, S. Tay “Real-time tracking, retrieval and gene expression analysis of migrating human T cells“ Lab Chip 2015, 15:1217. [DOI]
K. Gademann “Copy, Edit, and Paste: Natural Product Approaches to Biomaterials and Neuroengineering“ Acc. Chem. Res. 2015, 48:731. [DOI]
M. Lomora, I. A. Dinu, C. G. Palivan “Selective ion-permeable membranes by insertion of biopores into polymersomes“ Phys. Chem. 2015, 17:15538. [DOI]
R. S. Wagner, L. E. Kapinos, N. J. Marshall, M. Stewart, R. Y. Lim “Promiscuous binding of Karyopherinbeta1 modulates FG nucleoporin barrier function and expedites NTF2 transport kinetics“ Biophys. J. 2015, 108:918-27. [DOI]
J. M. Nunes, M. Mayer-Hartl, F. U. Hartl, D. J. Müller “Action of the Hsp70 chaperone system observed with single proteins“ Nat. Commun. 2015, 6:6307. [DOI] [More Information]
H. Miyatake-Ondozabal, E. Kaufmann, K. Gademann “Total Synthesis of the Protected Aglycon of Fidaxomicin (Tiacumicin B, Lipiarmycin A3)“ Angew. Chem. Int. Ed. 2015, 54:1933. [DOI]
B. Geering, M. Fussenegger “Synthetic immunology: modulating the human immune system“ Trends Biotechnol. 2015, 33:65-79. [DOI]
R. A. Kellogg, S. Tay “Noise facilitates transcriptional control under dynamic inputs“ Cell 2015, 160:381-92. [DOI]
M. Spulber, P. Baumann, J. Liu, C. G. Palivan “Ceria loaded nanoreactors: a nontoxic superantioxidant system with high stability and efficacy“ Nanoscale 2015, 7:1411. [DOI]
P. D. Bosshart, A. Engel, D. Fotiadis “High-Resolution Atomic Force Microscopy Imaging of Rhodopsin in Rod Outer Segment Disk Membranes“ Rhodopsin 2015:189-203. [DOI]
B. Dielacher, R. F. Tiefenauer, J. Junesch, J. Vörös “Iodide sensing via electrochemical etching of ultrathin gold films“ Nanotechnology 2015, 26:025202. [DOI]
T. Quinto, D. Haussinger, V. Kohler, T. R. Ward “Artificial metalloenzymes for the diastereoselective reduction of NAD(+) to NAD(2)H“ Org. Biomol. Chem. 2015, 13:357-60. [DOI]
N. Chuard, G. Gasparini, A. Roux, N. Sakai, S. Matile “Cell-penetrating poly(disulfide)s: the dependence of activity, depolymerization kinetics and intracellular localization on their length“ Org. Biomol. Chem. 2015, 13:64-7. [DOI]
E. Schönhofer, B. Bozic-Weber, C. J. Martin, E. C. Constable, C. E. Housecroft, J. A. Zampese “‘Surfaces-as-ligands, surfaces-as-complexes’ strategies for copper(I) dye-sensitized solar cells“ Dyes Pigm. 2015, 115:154. [DOI]
T. Schuster, M. Nussbaumer, P. Baumann, N. Bruns, W. Meier, A. Car “Polymeric particulates for subunit vaccine delivery“ Subunit Vaccine Delivery 2015:181-201. [DOI]
D. Alsteens, S. Tay, D. J. Müller “Toward high-throughput biomechanical phenotyping of single molecules“ Nat. Methods 2015, 12:45. [DOI]
G. Gunkel-Grabole, S. Sigg, M. Lomora, S. Lörcher, C. G. Palivan “Polymeric 3D nano-architectures for transport and delivery of therapeutically relevant biomacromolecules“ Biomater. Sci. 2015, 2:25. [DOI]
A. Najer, D. Wu, A. Bieri, F. Brand, C. G. Palivan, H. P. Beck, W. Meier “Nanomimics of Host Cell Membranes Block Invasion and Expose Invasive Malaria Parasites“ ACS Nano 2014, 8:12560. [DOI] [More Information]
F. Itel, M. Chami, A. Najer, S. Lörcher, D. Wu, I. A. Dinu “Molecular organization and dynamics in polymersome membranes: A lateral diffusion study“ Macromolecules 2014, 47:7588. [DOI]
T. Szikra, S. Trenholm, A. Drinnenberg, J. Jüttner, Z. Raics, K. Farrow, M. Biel, G. Awatramani, D. A. Clark, J. A. Sahel, R. Azeredo da Silveira, B. Roska “Rods in daylight act as relay cells for cone-driven horizontal cell–mediated surround inhibition“ Nat. Neurosci. 2014, 17:1728. [DOI]
K. Fujisawa, C. Beuchat, M. Humbert-Droz, A. Wilson, T. A. Wesolowski, J. Mareda, N. Sakai, S. Matile “Anion-π and cation-π interactions on the same surface“ Angew. Chem. Int. Ed. 2014, 53:11266-9. [DOI]
D. Steuerwald, S. M. Fruh, R. Griss, R. D. Lovchik, V. Vogel “Nanoshuttles propelled by motor proteins sequentially assemble molecular cargo in a microfluidic device“ Lab Chip 2014, 14:3729-38. [DOI]
F. N. Miros, G. Huang, Y. Zhao, N. Sakai, S. Matile “Coumarin synthesis on π-acidic surfaces“ Supramol. Chem. 2014, 27:303-09. [DOI]
K. Yonehara, B. Roska “Neuroscience: retinal projectome reveals organizing principles of the visual system“ Curr. Biol. 2014, 24:R833-5. [DOI]
H. Hayashi, A. Sobczuk, A. Bolag, N. Sakai, S. Matile “Antiparallel three-component gradients in double-channel surface architectures“ Chem. Sci. 2014, 5:4610-14. [DOI]
D. Wu, M. Spulber, F. Itel, M. Chami, T. Pfohl, C. G. Palivan, W. Meier “Effect of Molecular Parameters on the Architecture and Membrane Properties of 3D Assemblies of Amphiphilic Copolymers“ Macromolecules 2014, 47:5060-69. [DOI]
T. Cronin, L. H. Vandenberghe, P. Hantz, J. Juttner, A. Reimann, A. E. Kacsó, R. M. Huckfeldt, V. Busskamp, H. Kohler, P. S. Lagali, B. Roska, J. Bennett “Efficient transduction and optogenetic stimulation of retinal bipolar cells by a synthetic adeno-associated virus capsid and promoter“ EMBO Mol. Med. 2014, 6:1175. [DOI]
P. Baumann, M. Spulber, I. A. Dinu, C. G. Palivan “Cellular Trojan Horse Based Polymer Nanoreactors with Light-sensitive Activity“ J. Phys. Chem. B 2014, 118:9361. [DOI]
A. S. Chuong, M. L. Miri, V. Busskamp, G. A. C. Matthews, L. C. Acker, A. T. Sørensen, A. Young, N. C. Klapoetke, M. A. Henninger, S. B. Kodandaramaiah, M. Ogawa, S. B. Ramanlal, R. C. Bandler, B. D. Allen, C. R. Forest, B. Y. Chow, X. Han, Y. Lin, K. M. The, B. Roska, J. A. Cardin, E. S. Boyden “Noninvasive optical inhibition with a red-shifted microbial rhodopsin “ Nat. Neurosci. 2014, 17:1123. [DOI]
V. Busskamp, J. Krol, D. Nelidova, J. Daum, T. Szikra, B. Tsuda, J. Jüttner, K. Farrow, B. Gross Scherf, C. P. Patino Alvarez, C. Genoud, V. Sothilingam, N. Tanimoto, M. Stadler, M. Seeliger, M. Stoffel, W. Filipowicz, B. Roska “miRNAs 182 and 183 Are Necessary to Maintain Adult Cone Photoreceptor Outer Segments and Visual Function“ Neuron 2014, 83:586. [DOI]
R. A. Kellogg, R. Gomez-Sjoberg, A. A. Leyrat, S. Tay “High-throughput microfluidic single-cell analysis pipeline for studies of signaling dynamics“ Nat. Protoc. 2014, 9:1713-26. [DOI]
K. D. Schleicher, S. L. Dettmer, L. E. Kapinos, S. Pagliara, U. F. Keyser, S. Jeney, R. Y. H. Lim “Selective transport control on molecular velcro made from intrinsically disordered proteins“ Nat. Nanotechnol. 2014, 9:525. [DOI]
A. V. Jentzsch, S. Matile “Anion transport with halogen bonds“ Halogen Bonding I 2014:205-39. [DOI]

Displaying results 1 to 20 out of 44
« First ‹ Previous Page 1 Page 2 Page 3 Next › Last »

Project management Persons involved [grouplink]
Murielle Delley N.N.
Randall Platt Mariia Cherepkova
Maria Kuhn
Sushmita Poddar
Antonio Santinha
Florian Schmidt
Niels Weisbach

Platt group @D-BSSE

Florian Seebeck Alice Maurer

Seebeck group @UniBas

Ivan Martin
Yaakov Benenson
Manuele Giuseppe Muraro

Martin group @UniBas

Benenson group @D-BSSE

Catherine E. Housecroft
Cornelia G. Palivan
Viviana Maffeis
Myrto Kyropoulou
Dalin Wu

Housecroft group @UniBas

Palivan group @UniBas

Florian Seebeck
Petra Dittrich
N.N.

Seebeck group @UniBas

Dittrich group @D-BSSE

Bruno Correia N.N.
Martin Fussenegger N.N.

Meier group @UniBas

Fussenegger group @D-BSSE

Jonathan De Roo N.N.

De Roo group @UniBas

Marcel Mayor
Emanuel Lörtscher
N.N.

Mayor group @UniBas

Lörtscher group @IBM

Christof Sparr Daniel Moser
Felix Raps
Dragan Miladinov
Zlatko Joncev

Sparr group @UniBas

Marcel Mayor Marius Ciobanu
Tim Hohner
Gabriel Puebla-Hellman
David Vogel
Giulia Prone

Mayor group @UniBas

Konrad Tiefenbacher
Cornelia G. Palivan
N.N.

Tiefenbacher group @UniBas

Palivan group @UniBas

Sven Panke Daniel Gerngross
Lukas Huber
Markus Jeschek
Eirini Rousounelou
Peter Ruppen
Sanja Tunjic

Panke group @D-BSSE

Dimitrios Fotiadis Stephan Hirschi
Mirko Stauffer

Fotiadis group @UniBe

Botond Roska Jacek Krol
Magdalena Renner
Tamas Szikra

Roska group @FMI

Bruno Correia
Sai Reddy
Pablo Gainza

Correia group @EPFL

Reddy group @D-BSSE

Dimitrios Fotiadis
Daniel J. Müller
N.N.

Fotiadis group @UniBe

Meier group @UniBas

Müller group @D-BSSE

Effy Vayena Alessandro Blasimme
Renan Goncalves Leonel da Silva

Vayena group @ETHZ

Sai Reddy Theresa Pesch
Derek Mason
Jakub Kucharczyk

Reddy group @D-BSSE