Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Sci Adv ; 7(50): eabk0882, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34890224

ABSTRACT

Imaging molecular patterns in cells by fluorescence micro- or nanoscopy has the potential to relate collective molecular behavior to cellular function. However, spatial and spectroscopic resolution is fundamentally limited by motional blur caused by finite photon fluxes and photobleaching. At physiological temperatures, photochemical reactivity does not only limit imaging at multiple scales but is also toxic to biochemical reactions that maintain cellular organization. Here, we present cryoprotectant-free ultrarapid cryo-arrest directly on a multimodal fluorescence microscope that preserves the out-of-equilibrium molecular organization of living cells. This allows the imaging of dynamic processes before cryo-arrest in combination with precise molecular pattern determination at multiple scales within the same cells under cryo-arrest. We both experimentally and theoretically show that ultrarapid cryo-arrest overcomes the fundamental resolution barrier imposed by motional blur and photochemical reactivity, enabling observation of native molecular distributions and reaction patterns that are not resolvable at physiological temperatures.

2.
Methods Mol Biol ; 2262: 199-215, 2021.
Article in English | MEDLINE | ID: mdl-33977478

ABSTRACT

Reaction-diffusion simulations allow us to recapitulate experimentally observed behavior, e.g., from time series of fluorescent micrographs. This is essential to inform our intuitive understanding of the chemical and biophysical interaction of proteins in a cellular context as well as their role in reaction networks. This chapter aims to give a brief introduction to setting up reaction-diffusion simulations and applies these in silico techniques to take apart the spatial cycles that maintain Ras localization in the cell.


Subject(s)
Computer Simulation , Guanosine Triphosphate/metabolism , Models, Biological , Spatial Analysis , ras Proteins/metabolism , Diffusion , Humans
3.
Sci Signal ; 14(683)2021 05 18.
Article in English | MEDLINE | ID: mdl-34006609

ABSTRACT

Growth factor-dependent vesicular dynamics allow cells to regulate the spatial distribution of growth factor receptors and thereby their coupling to downstream signaling effectors that guide cellular responses. We found that the ErbB ligands epidermal growth factor (EGF) and heregulin (HRG) generated distinct spatiotemporal patterns of cognate receptor activities to activate distinct subcellular pools of the extracellular signal-regulated kinase (Erk). Sustained plasma membrane activity of the receptor tyrosine kinases ErbB2/ErbB3 signaled to Erk complexed with the scaffold protein KSR to promote promigratory EphA2 phosphorylation and cellular motility upon HRG stimulation. In contrast, receptor-saturating EGF stimuli caused proliferation-inducing transient activation of cytoplasmic Erk due to the rapid internalization of EGF receptors (EGFR or ErbB1) toward endosomes. Paradoxically, promigratory signaling mediated by Erk complexed to KSR was sustained at low EGF concentrations by vesicular recycling that maintained steady-state amounts of active, phosphorylated EGFR at the plasma membrane. Thus, the effect of ligand identity and concentration on determining ErbB vesicular dynamics constitutes a mechanism by which cells can transduce growth factor composition through spatially distinct Erk pools to enable functionally diverse cellular responses.


Subject(s)
Extracellular Signal-Regulated MAP Kinases , Receptor, ErbB-2 , Cell Movement , Epidermal Growth Factor/metabolism , Phosphorylation , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction
4.
Nat Commun ; 12(1): 1548, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33750780

ABSTRACT

Reconstituting artificial proto-cells capable of transducing extracellular signals into cytoskeletal changes can reveal fundamental principles of how non-equilibrium phenomena in cellular signal transduction affect morphogenesis. Here, we generated a Synthetic Morphogenic Membrane System (SynMMS) by encapsulating a dynamic microtubule (MT) aster and a light-inducible signaling system driven by GTP/ATP chemical potential into cell-sized liposomes. Responding to light cues in analogy to morphogens, this biomimetic design embodies basic principles of localized Rho-GTPase signal transduction that generate an intracellular MT-regulator signaling gradient. Light-induced signaling promotes membrane-deforming growth of MT-filaments by dynamically elevating the membrane-proximal tubulin concentration. The resulting membrane deformations enable recursive coupling of the MT-aster with the signaling system, which generates global self-organized morphologies that reorganize towards local external cues in dependence on prior shape. SynMMS thereby signifies a step towards bio-inspired engineering of self-organized cellular morphogenesis.


Subject(s)
Cues , Liposomes , Morphogenesis/physiology , Artificial Cells , Biophysical Phenomena , Cell Surface Extensions/physiology , Centrosome , Cytoskeleton/metabolism , Humans , Liposomes/chemistry , Microtubules/metabolism , Recombinant Proteins , Signal Transduction , Stathmin/metabolism , Synthetic Biology , Tubulin/metabolism , rho GTP-Binding Proteins/metabolism
5.
Annu Rev Cell Dev Biol ; 36: 359-383, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32692593

ABSTRACT

The proto-oncogenic epidermal growth factor (EGF) receptor (EGFR) is a tyrosine kinase whose sensitivity and response to growth factor signals that vary over time and space determine cellular behavior within a developing tissue. The molecular reorganization of the receptors on the plasma membrane and the enzyme-kinetic mechanisms of phosphorylation are key determinants that couple growth factor binding to EGFR signaling. To enable signal initiation and termination while simultaneously accounting for suppression of aberrant signaling, a coordinated coupling of EGFR kinase and protein tyrosine phosphatase activity is established through space by vesicular dynamics. The dynamical operation mode of this network enables not only time-varying growth factor sensing but also adaptation of the response depending on cellular context. By connecting spatially coupled enzymatic kinase/phosphatase processes and the corresponding dynamical systems description of the EGFR network, we elaborate on the general principles necessary for processing complex growth factor signals.


Subject(s)
ErbB Receptors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Biocatalysis , Cell Plasticity , ErbB Receptors/chemistry , Humans , Signal Transduction , Time Factors
6.
Cell Chem Biol ; 26(6): 842-851.e7, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30956149

ABSTRACT

Interference with the signaling activity of the N-myristoylated nonreceptor protein tyrosine kinase Src is considered a viable approach in anti-cancer drug discovery. However, ATP-competitive Src inhibitors have not reached the clinic yet and alternative approaches are in high demand. The UNC119A/B proteins bind the myristoylated N terminus of Src and thereby mediate energy-driven spatial cycles that maintain Src enrichment at the plasma membrane, which is critical for Src signaling activity. We describe the discovery of a potent and specific inhibitor of the UNC119-Src interaction with unprecedented chemotype. The inhibitor binds to UNC119 in cells, and induces redistribution of Src to endomembranes and reduction of activating Src autophosphorylation on Y419. UNC119 inhibition in Src-dependent colorectal cancer cells results in the specific reduction of cell growth and clonogenic potential. Our results demonstrate that small-molecule interference with the dynamics of the Src spatial cycle may provide an opportunity to impair oncogenic Src signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Small Molecule Libraries/pharmacology , src-Family Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Female , Humans , Male , Mice , Molecular Structure , Small Molecule Libraries/chemistry , src-Family Kinases/metabolism
7.
Int J Cancer ; 144(4): 767-776, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30194764

ABSTRACT

Ras proteins, most notably KRas, are prevalent oncogenes in human cancer. Plasma membrane localization and thereby signaling of KRas is regulated by the prenyl-binding protein PDEδ. Recently, we have reported the specific anti-proliferative effects of PDEδ inhibition in KRas-dependent human pancreatic ductal adenocarcinoma cell lines. Here, we investigated the proliferative dependence on the solubilizing activity of PDEδ of human colorectal cancer (CRC) cell lines with or without oncogenic KRas mutations. Our results show that genetic and pharmacologic interference with PDEδ specifically inhibits proliferation and survival of CRC cell lines harboring oncogenic KRas mutations whereas isogenic cell lines in which the KRas oncogene has been removed, or cell lines with oncogenic BRaf mutations or EGFR overexpression are not dependent on PDEδ. Pharmacological PDEδ inhibition is therefore a possible new avenue to target oncogenic KRas bearing CRC.


Subject(s)
Benzimidazoles/pharmacology , Cell Proliferation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 6/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RNA Interference
8.
Nat Commun ; 9(1): 3847, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242154

ABSTRACT

Epidermal growth factor receptor (EGFR) activation by growth factors (GFs) relies on dimerization and allosteric activation of its intrinsic kinase activity, resulting in trans-phosphorylation of tyrosines on its C-terminal tail. While structural and biochemical studies identified this EGF-induced allosteric activation, imaging collective EGFR activation in cells and molecular dynamics simulations pointed at additional catalytic EGFR activation mechanisms. To gain more insight into EGFR activation mechanisms in living cells, we develop a Förster resonance energy transfer (FRET)-based conformational EGFR indicator (CONEGI) using genetic code expansion that reports on conformational transitions in the EGFR activation loop. Comparing conformational transitions, self-association and auto-phosphorylation of CONEGI and its Y845F mutant reveals that Y845 phosphorylation induces a catalytically active conformation in EGFR monomers. This conformational transition depends on EGFR kinase activity and auto-phosphorylation on its C-terminal tail, generating a looped causality that leads to autocatalytic amplification of EGFR phosphorylation at low EGF dose.


Subject(s)
ErbB Receptors , Intercellular Signaling Peptides and Proteins , Protein Conformation , Dimerization , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Phosphorylation
9.
Redox Biol ; 19: 210-217, 2018 10.
Article in English | MEDLINE | ID: mdl-30176560

ABSTRACT

In order to overcome intercellular variability and thereby effectively assess signal propagation in biological networks it is imperative to simultaneously quantify multiple biological observables in single living cells. While fluorescent biosensors have been the tool of choice to monitor the dynamics of protein interaction and enzymatic activity, co-measuring more than two of them has proven challenging. In this work, we designed three spectrally separated anisotropy-based Förster Resonant Energy Transfer (FRET) biosensors to overcome this difficulty. We demonstrate this principle by monitoring the activation of extrinsic, intrinsic and effector caspases upon apoptotic stimulus. Together with modelling and simulations we show that time of maximum activity for each caspase can be derived from the anisotropy of the corresponding biosensor. Such measurements correlate relative activation times and refine existing models of biological signalling networks, providing valuable insight into signal propagation.


Subject(s)
Apoptosis , Caspases, Effector/analysis , Microscopy, Fluorescence/methods , Biosensing Techniques/methods , Caspases, Effector/metabolism , Enzyme Activation , Fluorescence Polarization/methods , Fluorescence Resonance Energy Transfer/methods , HeLa Cells , Humans , Signal Transduction
10.
Sci Signal ; 11(541)2018 07 31.
Article in English | MEDLINE | ID: mdl-30065026

ABSTRACT

The ability of cells to adapt their response to growth factors in relation to their environment is an essential aspect of tissue development and homeostasis. We found that signaling mediated by the Eph family of receptor tyrosine kinases from cell-cell contacts changed the cellular response to the growth factor EGF by modulating the vesicular trafficking of its receptor, EGFR. Eph receptor activation trapped EGFR in Rab5-positive early endosomes by inhibiting Akt-dependent vesicular recycling. By altering the spatial distribution of EGFR activity, EGF-promoted Akt signaling from the plasma membrane was suppressed, thereby inhibiting cell migration. In contrast, ERK signaling from endosomal EGFR was preserved to maintain a proliferative response to EGF stimulation. We also found that soluble extracellular signals engaging the G protein-coupled receptor Kiss1 (Kiss1R) similarly suppressed EGFR vesicular recycling to inhibit EGF-promoted migration. Eph or Kiss1R activation also suppressed EGF-promoted migration in Pten-/- mouse embryonic fibroblasts, which exhibit increased constitutive Akt activity, and in MDA-MB-231 triple-negative breast cancer cells, which overexpress EGFR. The cellular environment can thus generate context-dependent responses to EGF stimulation by modulating EGFR vesicular trafficking dynamics.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Cytoplasmic Vesicles/physiology , Epidermal Growth Factor/pharmacology , Receptors, Eph Family/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cells, Cultured , Endocytosis , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , HEK293 Cells , Humans , Mice , PTEN Phosphohydrolase/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Eph Family/genetics , Receptors, Kisspeptin-1/genetics , Receptors, Kisspeptin-1/metabolism , Signal Transduction
11.
Cell Syst ; 7(3): 295-309.e11, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30145116

ABSTRACT

The proto-oncogenic epidermal growth factor receptor (EGFR) is a tyrosine kinase whose sensitivity to growth factors and signal duration determines cellular behavior. We resolve how EGFR's response to epidermal growth factor (EGF) originates from dynamically established recursive interactions with spatially organized protein tyrosine phosphatases (PTPs). Reciprocal genetic PTP perturbations enabled identification of receptor-like PTPRG/J at the plasma membrane and ER-associated PTPN2 as the major EGFR dephosphorylating activities. Imaging spatial-temporal PTP reactivity revealed that vesicular trafficking establishes a spatially distributed negative feedback with PTPN2 that determines signal duration. On the other hand, single-cell dose-response analysis uncovered a reactive oxygen species-mediated toggle switch between autocatalytically activated monomeric EGFR and the tumor suppressor PTPRG that governs EGFR's sensitivity to EGF. Vesicular recycling of monomeric EGFR unifies the interactions with these PTPs on distinct membrane systems, dynamically generating a network architecture that can sense and respond to time-varying growth factor signals.


Subject(s)
Cell Membrane/metabolism , Cytoplasmic Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Computational Biology , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Feedback, Physiological , Humans , MCF-7 Cells , Microscopy, Confocal , Models, Theoretical , Phosphorylation , Protein Interaction Maps , Protein Transport , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Signal Transduction , Single-Cell Analysis
12.
J Physiol ; 596(7): 1227-1241, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29369356

ABSTRACT

KEY POINTS: Na+ conducting hypertonicity-induced cation channels (HICCs) are key players in the volume restoration of osmotically shrunken cells and, under isotonic conditions, considered as mediators of proliferation - thereby opposing apoptosis. In an siRNA screen of ion channels and transporters in HepG2 cells, with the regulatory volume increase (RVI) as read-out, δENaC, TRPM2 and TRPM5 were identified as HICCs. Subsequently, all permutations of these channels were tested in RVI and patch-clamp recordings and, at first sight, HICCs were found to operate in an independent mode. However, there was synergy in the siRNA perturbations of HICC currents. Accordingly, proximity ligation assays showed that δENaC was located in proximity to TRPM2 and TRPM5 suggesting a physical interaction. Furthermore, δENaC, TRPM2 and TRPM5 were identified as mediators of HepG2 proliferation - their silencing enhanced apoptosis. Our study defines the architecture of HICCs in human hepatocytes as well as their molecular functions. ABSTRACT: Hypertonicity-induced cation channels (HICCs) are a substantial element in the regulatory volume increase (RVI) of osmotically shrunken cells. Under isotonic conditions, they are key effectors in the volume gain preceding proliferation; HICC repression, in turn, significantly increases apoptosis rates. Despite these fundamental roles of HICCs in cell physiology, very little is known concerning the actual molecular architecture of these channels. Here, an siRNA screening of putative ion channels and transporters was performed, in HepG2 cells, with the velocity of RVI as the read-out; in this first run, δENaC, TRPM2 and TRPM5 could be identified as HICCs. In the second run, all permutations of these channels were tested in RVI and patch-clamp recordings, with special emphasis on the non-additivity and additivity of siRNAs - which would indicate molecular interactions or independent ways of channel functioning. At first sight, the HICCs in HepG2 cells appeared to operate rather independently. However, a proximity ligation assay revealed that δENaC was located in proximity to both TRPM2 and TRPM5. Furthermore, a clear synergy of HICC current knock-downs (KDs) was observed. δENaC, TRPM2 and TRPM5 were defined as mediators of HepG2 cell proliferation and their silencing increased the rates of apoptosis. This study provides a molecular characterization of the HICCs in human hepatocytes and of their role in RVI, cell proliferation and apoptosis.


Subject(s)
Apoptosis , Cell Proliferation , Epithelial Sodium Channels/metabolism , Hepatocytes/pathology , Muscle Hypertonia/physiopathology , TRPM Cation Channels/metabolism , Epithelial Sodium Channels/chemistry , Epithelial Sodium Channels/genetics , Hep G2 Cells , Hepatocytes/metabolism , Humans , RNA, Small Interfering , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics
13.
Nat Commun ; 8(1): 466, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28878211

ABSTRACT

Reactive oxygen species are produced transiently in response to cell stimuli, and function as second messengers that oxidize target proteins. Protein-tyrosine phosphatases are important reactive oxygen species targets, whose oxidation results in rapid, reversible, catalytic inactivation. Despite increasing evidence for the importance of protein-tyrosine phosphatase oxidation in signal transduction, the cell biological details of reactive oxygen species-catalyzed protein-tyrosine phosphatase inactivation have remained largely unclear, due to our inability to visualize protein-tyrosine phosphatase oxidation in cells. By combining proximity ligation assay with chemical labeling of cysteine residues in the sulfenic acid state, we visualize oxidized Src homology 2 domain-containing protein-tyrosine phosphatase 2 (SHP2). We find that platelet-derived growth factor evokes transient oxidation on or close to RAB5+/ early endosome antigen 1- endosomes. SHP2 oxidation requires NADPH oxidases (NOXs), and oxidized SHP2 co-localizes with platelet-derived growth factor receptor and NOX1/4. Our data demonstrate spatially and temporally limited protein oxidation within cells, and suggest that platelet-derived growth factor-dependent "redoxosomes," contribute to proper signal transduction.Protein-tyrosine phosphatases (PTPs) are thought to be major targets of receptor-activated reactive oxygen species (ROS). Here the authors describe a method that allows the localized visualization of oxidized intermediates of PTPs inside cells during signaling, and provide support for the "redoxosome" model.


Subject(s)
Microscopy, Confocal/methods , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction , Time-Lapse Imaging/methods , 3T3 Cells , Animals , Cells, Cultured , Cysteine/genetics , Cysteine/metabolism , Hep G2 Cells , Humans , Immunoblotting , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Oxidation-Reduction/drug effects , Platelet-Derived Growth Factor/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Reactive Oxygen Species/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism
14.
Nat Commun ; 8(1): 114, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740133

ABSTRACT

The peripheral membrane proto-oncogene Src family protein tyrosine kinases relay growth factor signals to the cytoplasm of mammalian cells. We unravel the spatial cycles of solubilisation, trapping on perinuclear membrane compartments and vesicular transport that counter entropic equilibration to endomembranes for maintaining the enrichment and activity of Src family protein tyrosine kinases at the plasma membrane. The solubilising factor UNC119 sequesters myristoylated Src family protein tyrosine kinases from the cytoplasm, enhancing their diffusion to effectively release Src family protein tyrosine kinases on the recycling endosome by localised Arl2/3 activity. Src is then trapped on the recycling endosome via electrostatic interactions, whereas Fyn is quickly released to be kinetically trapped on the Golgi by palmitoyl acyl-transferase activity. Vesicular trafficking from these compartments restores enrichment of the Src family protein tyrosine kinases to the plasma membrane. Interference with these spatial cycles by UNC119 knockdown disrupts Src family protein tyrosine kinase localisation and signalling activity, indicating that UNC119 could be a drug target to affect oncogenic Src family protein tyrosine kinase signalling.The peripheral membrane proto-oncogene Src family protein tyrosine kinases (SFKs) transmit growth factor signals to the cytoplasm. Here the authors show that the solubilising factor UNC119 sequesters myristoylated SFKs to maintain its enrichment at the plasma membrane to enable signal transduction.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/metabolism , Signal Transduction , src-Family Kinases/metabolism , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Endosomes/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Golgi Apparatus/metabolism , HT29 Cells , HeLa Cells , Humans , Immunoblotting , Microscopy, Confocal , Models, Biological , Myristic Acid/metabolism , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , RNA Interference , Solubility , src-Family Kinases/genetics
15.
Bio Protoc ; 7(8)2017 Apr 20.
Article in English | MEDLINE | ID: mdl-28580376

ABSTRACT

Fluorescence live-cell imaging by single molecule localization microscopy (SMLM) or fluorescence lifetime imaging microscopy (FLIM) in principle allows for the spatio-temporal observation of molecular patterns in individual, living cells. However, the dynamics of molecules within cells hamper their precise observation. We present here a detailed protocol for consecutive cycles of reversible cryo-arrest of living cells on a microscope that allows for a precise determination of the evolution of molecular patterns within individual living cells. The usefulness of this approach has been demonstrated by observing ligand-induced clustering of receptor tyrosine kinases as well as their activity patterns by SMLM and FLIM (Masip et al., 2016).

16.
Angew Chem Int Ed Engl ; 56(9): 2423-2428, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28106325

ABSTRACT

Small-molecule inhibition of the interaction between the KRas oncoprotein and the chaperone PDE6δ impairs KRas spatial organization and signaling in cells. However, despite potent binding in vitro (KD <10 nm), interference with Ras signaling and growth inhibition require 5-20 µm compound concentrations. We demonstrate that these findings can be explained by fast release of high-affinity inhibitors from PDE6δ by the release factor Arl2. This limitation is overcome by novel highly selective inhibitors that bind to PDE6δ with up to 7 hydrogen bonds, resulting in picomolar affinity. Their release by Arl2 is greatly decreased, and representative compounds selectively inhibit growth of KRas mutated and -dependent cells with the highest activity recorded yet. Our findings indicate that very potent inhibitors of the KRas-PDE6δ interaction may impair the growth of tumors driven by oncogenic KRas.

17.
Nat Methods ; 13(8): 665-672, 2016 08.
Article in English | MEDLINE | ID: mdl-27400419

ABSTRACT

The dynamics of molecules in living cells hampers precise imaging of molecular patterns by functional and super-resolution microscopy. We developed a method that circumvents lethal chemical fixation and allows on-stage cryo-arrest for consecutive imaging of molecular patterns within the same living, but arrested, cells. The reversibility of consecutive cryo-arrests was demonstrated by the high survival rate of different cell lines and by intact growth factor signaling that was not perturbed by stress response. Reversible cryo-arrest was applied to study the evolution of ligand-induced receptor tyrosine kinase activation at different scales. The nanoscale clustering of epidermal growth factor receptor (EGFR) in the plasma membrane was assessed by single-molecule localization microscopy, and endosomal microscale activity patterns of ephrin receptor A2 (EphA2) were assessed by fluorescence lifetime imaging microscopy. Reversible cryo-arrest allows the precise determination of molecular patterns while conserving the dynamic capabilities of living cells.


Subject(s)
Cold Temperature , Cryoprotective Agents/chemistry , ErbB Receptors/metabolism , Microscopy, Fluorescence/methods , Molecular Imaging/methods , Receptor, EphA2/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , HeLa Cells , Humans , Phosphorylation , Signal Transduction
18.
Nat Commun ; 7: 11360, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-27094677

ABSTRACT

The prenyl-binding protein PDEδ is crucial for the plasma membrane localization of prenylated Ras. Recently, we have reported that the small-molecule Deltarasin binds to the prenyl-binding pocket of PDEδ, and impairs Ras enrichment at the plasma membrane, thereby affecting the proliferation of KRas-dependent human pancreatic ductal adenocarcinoma cell lines. Here, using structure-based compound design, we have now identified pyrazolopyridazinones as a novel, unrelated chemotype that binds to the prenyl-binding pocket of PDEδ with high affinity, thereby displacing prenylated Ras proteins in cells. Our results show that the new PDEδ inhibitor, named Deltazinone 1, is highly selective, exhibits less unspecific cytotoxicity than the previously reported Deltarasin and demonstrates a high correlation with the phenotypic effect of PDEδ knockdown in a set of human pancreatic cancer cell lines.


Subject(s)
Antineoplastic Agents/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 6/chemistry , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Phosphodiesterase Inhibitors/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , Pyrazines/chemistry , Pyrazoles/chemistry , Small Molecule Libraries/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 6/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression , Humans , Molecular Docking Simulation , Pancreatic Ducts/drug effects , Pancreatic Ducts/metabolism , Pancreatic Ducts/pathology , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/pharmacology , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrazines/chemical synthesis , Pyrazines/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology
19.
Biophys J ; 110(4): 840-9, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26541066

ABSTRACT

Complex living systems such as mammalian cells can be arrested in a solid phase by ultrarapid cooling. This allows for precise observation of cellular structures as well as cryopreservation of cells. The state of water, the main constituent of biological samples, is crucial for the success of cryogenic applications. Water exhibits many different solid states. If it is cooled extremely rapidly, liquid water turns into amorphous ice, also called vitreous water, a glassy and amorphous solid. For cryo-preservation, the vitrification of cells is believed to be mandatory for cell survival after freezing. Intracellular ice crystallization is assumed to be lethal, but experimental data on the state of water during cryopreservation are lacking. To better understand the water conditions in cells subjected to freezing protocols, we chose to directly analyze their subcellular water states by cryo-electron microscopy and tomography, cryoelectron diffraction, and x-ray diffraction both in the cryofixed state and after warming to different temperatures. By correlating the survival rates of cells with their respective water states during cryopreservation, we found that survival is less dependent on ice-crystal formation than expected. Using high-resolution cryo-imaging, we were able to directly show that cells tolerate crystallization of extra- and intracellular water. However, if warming is too slow, many small ice crystals will recrystallize into fewer but bigger crystals, which is lethal. The applied cryoprotective agents determine which crystal size is tolerable. This suggests that cryoprotectants can act by inhibiting crystallization or recrystallization, but they also increase the tolerance toward ice-crystal growth.


Subject(s)
Cryopreservation/methods , Ice , Cell Survival , Cryoelectron Microscopy , Crystallization , HeLa Cells , Humans , X-Ray Diffraction
20.
J Physiol ; 594(6): 1663-76, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26593308

ABSTRACT

Slow cooling leads to a passive dehydration of cells, whereas rehydration during warming reflects the active regain of functionality. The ability to modulate such an energy demanding process could be instrumental in optimizing the cryo-arrest of living systems. In the present study, various levels of hypertonic stress were used to disturb the water content of cells and to define the energy profiles of aquaporins and (Na(+) conducting) cation channels during rehydration. Na(+) import was found to be the rate-limiting step in water restoration, whereas aquaporins merely played a permissive role. Indeed, regulated Na(+) import was increased 2-fold following cryo-arrests, thus facilitating the osmotic rehydration of cells. Freezing temperatures increased cell viscosity with a remarkable hysteresis and viscosity was a trigger of cation channels. The peptide hormone vasopressin was a further activator of channels, increasing the viability of post-cryo cells considerably. Hence, the hormone opens the path for a novel class of cryo-protectants with an intrinsic biological activity.


Subject(s)
Adaptation, Physiological , Cell Cycle Checkpoints , Cold-Shock Response , Freezing , Osmotic Pressure , Aquaporins/metabolism , Cell Survival/drug effects , HeLa Cells , Hep G2 Cells , Humans , Vasopressins/pharmacology , Viscosity , Voltage-Gated Sodium Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...