Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Lipids Health Dis ; 23(1): 81, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38509584

ABSTRACT

BACKGROUND: Obesity is associated with extensive white adipose tissue (WAT) expansion and remodeling. Healthy WAT expansion contributes to the maintenance of energy balance in the liver, thereby ameliorating obesity-related hepatic steatosis. Tissue-resident mesenchymal stromal cell populations, including PDGFRß + perivascular cells, are increasingly recognized pivotal as determinants of the manner in which WAT expands. However, the full array of regulatory factors controlling WAT stromal cell functions remains to be fully elucidated. Hypoxia-inducible factors (HIFs) are critical regulators in WAT stromal cell populations such as adipocyte precursor cells (APCs). It is revealed that HIF1α activation within PDGFRß + stromal cells results in the suppression of de novo adipogenesis and the promotion of a pro-fibrogenic cellular program in obese animals. However, the role of HIF2α in PDGFRß + cells remains undetermined in vivo. METHODS: New genetic models were employed in which HIF1α (encoded by the Hif1a gene) and HIF2α (encoded by the Epas1 gene) are selectively inactivated in PDGFRß + cells in an inducible manner using tamoxifen (TAM). With these models, both in vitro and in vivo functional analysis of PDGFRß + cells lacking HIF proteins were performed. Additionally, comprehensive metabolic phenotyping in diet-induced mouse models were performed to investigate the roles of PDGFRß + cell HIF proteins in WAT remodeling, liver energy balance and systemic metabolism. RESULTS: Unlike HIF1α inactivation, the new findings in this study suggest that inducible ablation of HIF2α in PDGFRß + cells does not cause apparent effects on WAT expansion induced by obesogenic diet. The adipogenic ability of PDGFRß + APCs is not significantly altered by genetic HIF2α ablation. Moreover, no difference of key parameters associated with healthy WAT remodeling such as improvements of WAT insulin sensitivity, reduction in metabolic inflammation, as well as changes in liver fat accumulation or systemic glucose metabolism, is detected in PDGFRß + cell Epas1-deficient mice. CONCLUSION: The new findings in this study support that, in contrast to HIF1α, PDGFRß + cell HIF2α appears dispensable for WAT metabolic remodeling and the resulting effects on liver metabolic homeostasis in diet-induced obesity, underscoring the isoform-specific roles of HIFα proteins in the regulation of adipose tissue biology.


Subject(s)
Adipose Tissue, White , Basic Helix-Loop-Helix Transcription Factors , Obesity , Animals , Mice , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Lipids , Liver/metabolism , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism
2.
Chem Sci ; 14(7): 1687-1695, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36819876

ABSTRACT

Phosphatidylethanolamine metabolism plays essential roles in eukaryotic cells but has not been completely investigated due to its complexity. This is because lipid species, unlike proteins or nucleic acids, cannot be easily manipulated at the single molecule level or controlled with subcellular resolution, two of the key factors toward understanding their functions. Here, we use the organelle-targeting photoactivation method to study PE metabolism in living cells with a high spatiotemporal resolution. Containing predefined PE structures, probes which can be selectively introduced into the ER or mitochondria were designed to compare their metabolic products according to their subcellular localization. We combined photo-uncaging with dual stable isotopic labeling to track PE metabolism in living cells by mass spectrometry analysis. Our results reveal that both mitochondria- and ER-released PE participate in phospholipid remodeling, and that PE methylation can be detected only under particular conditions. Thus, our method provides a framework to study phospholipid metabolism at subcellular resolution.

3.
Commun Biol ; 5(1): 736, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35869234

ABSTRACT

Precursor messenger RNA splicing is a highly regulated process, mediated by a complex RNA-protein machinery, the spliceosome, that encompasses several hundred proteins and five small nuclear RNAs in humans. Emerging evidence suggests that the spatial organization of splicing factors and their spatio-temporal dynamics participate in the regulation of splicing. So far, methods to manipulate the spatial distribution of splicing factors in a temporally defined manner in living cells are missing. Here, we describe such an approach that takes advantage of a reversible chemical dimerizer, and outline the requirements for efficient, reversible re-localization of splicing factors to selected sub-nuclear compartments. In a proof-of-principle study, the partial re-localization of the PRPF38A protein to the nuclear lamina in HEK293T cells induced a moderate increase in intron retention. Our approach allows fast and reversible re-localization of splicing factors, has few side effects and can be applied to many splicing factors by fusion of a protein tag through genome engineering. Apart from the systematic analysis of the spatio-temporal aspects of splicing regulation, the approach has a large potential for the fast induction and reversal of splicing switches and can reveal mechanisms of splicing regulation in native nuclear environments.


Subject(s)
Nuclear Lamina , Spliceosomes , HEK293 Cells , Humans , Nuclear Lamina/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism
4.
ACS Chem Biol ; 17(6): 1485-1494, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35667650

ABSTRACT

Lipid metabolism is spatiotemporally regulated within cells, yet intervention into lipid functions at subcellular resolution remains difficult. Here, we report a method that enables site-specific release of sphingolipids and cholesterol inside the vacuole in Saccharomyces cerevisiae. Using this approach, we monitored real-time sphingolipid metabolic flux out of the vacuole by mass spectrometry and found that the endoplasmic reticulum-vacuole-tethering protein Mdm1 facilitated the metabolism of sphingoid bases into ceramides. In addition, we showed that cholesterol, once delivered into yeast using our method, could restore cell proliferation induced by ergosterol deprivation, overcoming the previously described sterol-uptake barrier under aerobic conditions. Together, these data define a new way to study intracellular lipid metabolism and transport from the vacuole in yeast.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Cholesterol/metabolism , Intermediate Filament Proteins/metabolism , Lipid Metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sphingolipids/chemistry , Sphingolipids/metabolism , Vacuoles/metabolism
5.
Chimia (Aarau) ; 75(12): 1012-1016, 2021 Dec 22.
Article in English | MEDLINE | ID: mdl-34920769

ABSTRACT

Lipids are important cellular components providing many essential functions. To fulfill these various functions evolution has selected for a diverse set of lipids and this diversity is seen at the organismal, cellular and subcellular level. Understanding how cells maintain this complex lipid organization is a very challenging problem, which for lipids, is not easily addressed using biochemical and genetic techniques. Therefore, chemical tools have an important role to play in our quest to understand the complexities of lipid metabolism. Here we discuss new chemical tools to study lipids, their distribution and metabolism with increased spatial and temporal resolution.


Subject(s)
Lipid Metabolism , Lipids , Biology
6.
J Am Chem Soc ; 143(10): 3665-3670, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33684293

ABSTRACT

Protein trafficking and protein-protein interactions (PPIs) are central to regulatory processes in cells. Induced dimerization systems have been developed to control PPIs and regulate protein trafficking (localization) or interactions. Chemically induced dimerization (CID) has proven to be a robust approach to control protein interactions and localization. The most recent embodiment of this technology relies on CID conjugates that react with a self-labeling protein on one side and a photocaged ligand on the other side to provide spatiotemporal control of the interaction with the protein of interest. Advancing this technology further is limited by the light delivery problem and the phototoxicity of intense irradiation necessary to achieve photouncaging. Herein, we designed a novel chemically induced dimerization system that was triggered by bioluminescence, instead of external light. Protein dimerization showed fast kinetics and was validated by an induced change of localization of a target protein (to and from the nucleus or plasma membrane) upon trigger. The technology was used transiently to activate the phosphatidylinositol 3-kinase (PI3K)/mTOR pathway and measure the impact on lipid synthesis/metabolism, assessed by lipidomics.


Subject(s)
Luciferases/metabolism , Proteins/metabolism , Dimerization , Protein Binding
7.
ACS Chem Biol ; 16(3): 452-456, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33586946

ABSTRACT

We report short ceramide analogs that can be activated with light and further functionalized using azide-alkyne click chemistry. These molecules, termed scaCers, exhibit increased cell permeability compared to their long-chain analogs as demonstrated using mass spectrometry and imaging. Notably, scaCers enable optical control of apoptosis, which is not observed with long-chain variants. Additionally, they function as photoswitchable substrates for sphingomyelin synthase 2 (SMS2), exhibiting inverted light-dependence compared to their extended analogs.


Subject(s)
Apoptosis/radiation effects , Ceramides/chemistry , Photosensitizing Agents/chemistry , Alkynes/chemistry , Azides/chemistry , Cell Membrane Permeability , Ceramides/metabolism , Click Chemistry , HeLa Cells , Humans , Photochemical Processes , Structure-Activity Relationship , Transferases (Other Substituted Phosphate Groups)/metabolism
8.
Curr Biol ; 30(19): 3775-3787.e7, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32857977

ABSTRACT

Sphingolipids play important roles in physiology and cell biology, but a systematic examination of their functions is lacking. We performed a genome-wide CRISPRi screen in sphingolipid-depleted human cells and identified hypersensitive mutants in genes of membrane trafficking and lipid biosynthesis, including ether lipid synthesis. Systematic lipidomic analysis showed a coordinate regulation of ether lipids with sphingolipids, suggesting an adaptation and functional compensation. Biophysical experiments on model membranes show common properties of these structurally diverse lipids that also share a known function as glycosylphosphatidylinositol (GPI) anchors in different kingdoms of life. Molecular dynamics simulations show a selective enrichment of ether phosphatidylcholine around p24 proteins, which are receptors for the export of GPI-anchored proteins and have been shown to bind a specific sphingomyelin species. Our results support a model of convergent evolution of proteins and lipids, based on their physico-chemical properties, to regulate GPI-anchored protein transport and maintain homeostasis in the early secretory pathway.


Subject(s)
Phospholipid Ethers/metabolism , Secretory Pathway/physiology , Sphingolipids/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Ether/analysis , Ether/metabolism , Glycosylphosphatidylinositols/analysis , Glycosylphosphatidylinositols/metabolism , Humans , Lipids/biosynthesis , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Mice , Protein Transport/physiology , Sphingolipids/physiology
9.
Angew Chem Int Ed Engl ; 58(45): 16033-16037, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31478317

ABSTRACT

Bioluminescence resonance energy transfer (BRET) has been widely used for studying dynamic processes in biological systems such as protein-protein interactions and other signaling events. Aside from acting as a reporter, BRET can also turn on functions in living systems. Herein, we report the application of BRET to performing a biorthogonal reaction in living cells; namely, releasing functional molecules through energy transfer to a coumarin molecule, a process termed bioluminolysis. An efficient BRET from Nanoluc-Halotag chimera protein (H-Luc) to a coumarin substrate yields the excited state of coumarin, which in turn triggers hydrolysis to uncage a target molecule. Compared to the conventional methods, this novel uncaging system requires no external light source and shows fast kinetics (t1/2 <2 min). We applied this BRET uncaging system to release a potent kinase inhibitor, ibrutinib, in living cells, highlighting its broad utility in controlling the supply of bioactive small molecules in vivo.


Subject(s)
Coumarins/metabolism , Fluorescence Resonance Energy Transfer/methods , Luciferases/metabolism , Luminescent Measurements/methods , Luminescent Proteins/metabolism , Pyrazoles/metabolism , Pyrimidines/metabolism , Adenine/analogs & derivatives , HeLa Cells , Humans , Piperidines , Protein Binding
10.
Nat Chem Biol ; 15(6): 623-631, 2019 06.
Article in English | MEDLINE | ID: mdl-31036923

ABSTRACT

Sphingosine-1-phosphate (S1P) plays important roles as a signaling lipid in a variety of physiological and pathophysiological processes. S1P signals via a family of G-protein-coupled receptors (GPCRs) (S1P1-5) and intracellular targets. Here, we report on photoswitchable analogs of S1P and its precursor sphingosine, respectively termed PhotoS1P and PhotoSph. PhotoS1P enables optical control of S1P1-3, shown through electrophysiology and Ca2+ mobilization assays. We evaluated PhotoS1P in vivo, where it reversibly controlled S1P3-dependent pain hypersensitivity in mice. The hypersensitivity induced by PhotoS1P is comparable to that induced by S1P. PhotoS1P is uniquely suited for the study of S1P biology in cultured cells and in vivo because it exhibits prolonged metabolic stability compared to the rapidly metabolized S1P. Using lipid mass spectrometry analysis, we constructed a metabolic map of PhotoS1P and PhotoSph. The formation of these photoswitchable lipids was found to be light dependent, providing a novel approach to optically probe sphingolipid biology.


Subject(s)
Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Lysophospholipids/chemistry , Mice , Models, Molecular , Molecular Structure , Optical Imaging , Photochemical Processes , Sphingosine/chemistry , Sphingosine/metabolism
11.
Chem Sci ; 10(8): 2253-2258, 2019 Feb 28.
Article in English | MEDLINE | ID: mdl-30881649

ABSTRACT

Lipids are essential components of eukaryotic cell membranes and play crucial roles in cellular signaling and metabolism. While increasing evidence shows that the activities of lipids are dependent upon subcellular localization, tools to study local lipid metabolism and signaling are limited. Herein, we report an approach that enabled us to selectively deliver photo-caged lipids into lysosomes and thereafter to quickly release the lipid molecules by illumination. On combining this method with genetic techniques and lipidomics, we were able to investigate the localization-dependent metabolism of an important intermediate of sphingolipid metabolism, sphingosine. Our data reveal a distinct metabolic pattern of lysosomal sphingosine. In general, this method has the potential to serve as a platform to study lysosomal metabolism and signaling of various lipids and metabolites in living cells.

12.
Elife ; 72018 01 29.
Article in English | MEDLINE | ID: mdl-29376826

ABSTRACT

Photoactivation ('uncaging') is a powerful approach for releasing bioactive small-molecules in living cells. Current uncaging methods are limited by the random distribution of caged molecules within cells. We have developed a mitochondria-specific photoactivation method, which permitted us to release free sphingosine inside mitochondria and thereafter monitor local sphingosine metabolism by lipidomics. Our results indicate that sphingosine was quickly phosphorylated into sphingosine 1-phosphate (S1P) driven by sphingosine kinases. In time-course studies, the mitochondria-specific uncaged sphingosine demonstrated distinct metabolic patterns compared to globally-released sphingosine, and did not induce calcium spikes. Our data provide direct evidence that sphingolipid metabolism and signaling are highly dependent on the subcellular location and opens up new possibilities to study the effects of lipid localization on signaling and metabolic fate.


Subject(s)
Cytological Techniques/methods , Lysophospholipids/metabolism , Mitochondria/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Animals , Cells, Cultured , Light , Mice , Mitochondria/radiation effects , Phosphorylation
13.
Chem Sci ; 8(5): 3676-3686, 2017 May 01.
Article in English | MEDLINE | ID: mdl-30155209

ABSTRACT

Sphingolipids are bio-active metabolites that show structural diversity among eukaryotes. They are essential for growth of all eukaryotic cells but when produced in an uncontrolled manner can lead to cell death and pathologies including auto-immune reactions, cancer, diabetes and neurodegeneration. Caenorhabditis elegans is an important genetic model organism both to find new drug-targets against parasitic nematodes and to study the conserved roles of sphingolipids in animals like their essential functions in very basic cellular processes ranging from maintenance of cell polarity and mitochondrial repair to growth and survival. C. elegans produces sphingoid bases which are structurally distinct from those of other animals as both iso- and anteiso-branched species have been reported. Using metabolic labeling we show that most worm sphingoid bases are iso-branched. We have synthesized the nematode-specific C17 iso-branched sphinganine and its 1-deoxy analogue and could show that both the iso-branch and the 1-hydroxyl group are essential to form functional nematode sphingolipids which are needed to maintain intestinal function. The organism specificity was examined by complementation experiments in Saccharomyces cerevisiae yeast cells lacking sphingoid base synthesis. We found that iso-branched sphingoid base did not support growth of mutant cells and was toxic to wild type yeast. 1-Deoxy sphingolipids have been linked to the hereditary disease HSAN1A and other metabolic disorders including diabetes. We found that in C. elegans the 1-deoxy analogue cannot rescue the intestinal phenotype caused by sphingoid base depletion. In fact, in wild-type animals with normal sphingoid base biosynthesis, exogenous 1-deoxy analogue had a disruptive effect on apical cytoskeletal organization of intestinal cells indicating that atypical bases can interfere with normal sphingolipid function.

14.
Chembiochem ; 17(22): 2118-2122, 2016 Nov 17.
Article in English | MEDLINE | ID: mdl-27782351

ABSTRACT

Selection of a specific neural stem/progenitor cells (NSPCs) has attracted broad attention in regenerative medicine for neurological disorders. Here, we report a fluorescent probe, CDg13, and its application for isolating strong neurogenic NSPCs. In comparison to the NSPCs isolated by other biomarkers, CDg13-stained NSPCs showed higher capability to differentiate into neurons. Target identification revealed that the fluorescence intensity of the probe within cells is inversely proportional to the expression levels of mouse and human Abcg2 transporters. These findings suggest that low Abcg2 expression is a biomarker for neurogenic NSPCs in mouse brain. Furthermore, CDg13 can be used to isolate Abcg2low cells from heterogeneous cell populations.


Subject(s)
Benzamides/chemistry , Fluorescent Dyes/chemistry , Neural Stem Cells/metabolism , Neurons/metabolism , Xanthenes/chemistry , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Benzamides/metabolism , Biomarkers/metabolism , Brain/cytology , Brain/metabolism , Cell Differentiation , Cells, Cultured , Embryo, Mammalian/cytology , Humans , Mice , Microscopy, Fluorescence , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neural Stem Cells/chemistry , Neural Stem Cells/cytology , Neurons/cytology , Propionates/chemistry , Propionates/metabolism , Quinolines/chemistry , Quinolines/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Xanthenes/metabolism
15.
Nat Commun ; 6: 10056, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26686736

ABSTRACT

Photoactivation of caged biomolecules has become a powerful approach to study cellular signalling events. Here we report a method for anchoring and uncaging biomolecules exclusively at the outer leaflet of the plasma membrane by employing a photocleavable, sulfonated coumarin derivative. The novel caging group allows quantifying the reaction progress and efficiency of uncaging reactions in a live-cell microscopy setup, thereby greatly improving the control of uncaging experiments. We synthesized arachidonic acid derivatives bearing the new negatively charged or a neutral, membrane-permeant coumarin caging group to locally induce signalling either at the plasma membrane or on internal membranes in ß-cells and brain slices derived from C57B1/6 mice. Uncaging at the plasma membrane triggers a strong enhancement of calcium oscillations in ß-cells and a pronounced potentiation of synaptic transmission while uncaging inside cells blocks calcium oscillations in ß-cells and causes a more transient effect on neuronal transmission, respectively. The precise subcellular site of arachidonic acid release is therefore crucial for signalling outcome in two independent systems.


Subject(s)
Arachidonic Acid/metabolism , Cell Membrane/metabolism , Animals , Arachidonic Acid/chemistry , Calcium/metabolism , Calcium Signaling/radiation effects , Cell Membrane/radiation effects , Coumarins/chemistry , Coumarins/metabolism , HeLa Cells , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/radiation effects , Light , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/radiation effects
16.
Bioorg Med Chem ; 23(12): 2868-76, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26004322

ABSTRACT

Most cellular processes are driven by simple biochemical mechanisms such as protein and lipid phosphorylation, but the sum of all these conversions is exceedingly complex. Hence, intuition alone is not enough to discern the underlying mechanisms in the light of experimental data. Toward this end, mathematical models provide a conceptual and numerical framework to formally evaluate the plausibility of biochemical processes. To illustrate the use of these models, here we built a mechanistic computational model of PI3K (phosphatidylinositol 3-kinase) activity, to determine the kinetics of lipid metabolizing enzymes in single cells. The model is trained to data generated upon perturbation with a reversible small-molecule based chemical dimerization system that allows for the very rapid manipulation of the PIP3 (phosphatidylinositol 3,4,5-trisphosphate) signaling pathway, and monitored with live-cell microscopy. We find that the rapid relaxation system used in this work decreased the uncertainty of estimating kinetic parameters compared to methods based on in vitro assays. We also examined the use of Bayesian parameter inference and how the use of such a probabilistic method gives information on the kinetics of PI3K and PTEN activity.


Subject(s)
PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Signal Transduction , Animals , Bayes Theorem , HeLa Cells , Humans , Kinetics , Models, Biological , Signal Transduction/drug effects , Small Molecule Libraries/metabolism
17.
Bioorg Med Chem ; 23(12): 2862-7, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25840797

ABSTRACT

Chemical dimerizers are powerful non-invasive tools for bringing molecules together inside intact cells. We recently introduced a rapidly reversible chemical dimerizer system which enables transient translocation of enzymes to and from the plasma membrane (PM). Here we have applied this system to transiently activate phosphatidylinositol 4,5-bisphosphate (PIP2) breakdown at the PM via translocation of phosphoinositide 5-phosphatase (5Ptase). We found that the PIP2 sensor phospholipase C-δ PH domain (PLCδ-PH) is released from the PM upon addition of the reversible chemical dimerizer rCD1. By outcompeting rCD1, rapid release of the 5Ptase from the PM is followed by PIP2 recovery. This permits the observation of the PIP2-dependent clathrin assembly at the PM.


Subject(s)
Cell Membrane/metabolism , Clathrin/metabolism , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Transport/drug effects , Cell Line , Cell Membrane/drug effects , Dimerization , Endocytosis/drug effects , Humans , Phosphatidylinositols/chemistry
18.
Chembiochem ; 15(12): 1765-8, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25045107

ABSTRACT

Copper-free click chemistry is currently the most promising and most rapidly developing technology for performing tailored chemical reactions inside intact living cells and animals. Its potential is particularly intensely explored in the field of live cell imaging, for both proteins and metabolites. Here we expand the application spectrum of click reactions to the chemical crosslinking of two proteins of choice in living cells. By combining strain-promoted Diels-Alder cycloaddition with FlAsH-based labeling of peptidic tetracysteine motifs, we developed the membrane-permeating reversible crosslinker T-CrAsH. We demonstrate the feasibility of the method both in vitro and inside cells. The biggest advantage of this new tool is the small size of the crosslinkable groups; this significantly decreases the risk of functional interference.


Subject(s)
Cross-Linking Reagents/chemistry , Proteins/chemistry , Amino Acids/chemistry , Click Chemistry , Cross-Linking Reagents/chemical synthesis , HEK293 Cells , HeLa Cells , Humans , Molecular Structure , Proteins/metabolism
19.
Angew Chem Int Ed Engl ; 53(26): 6720-3, 2014 Jun 23.
Article in English | MEDLINE | ID: mdl-24841150

ABSTRACT

Chemical dimerizers are powerful tools for non-invasive manipulation of enzyme activities in intact cells. Here we introduce the first rapidly reversible small-molecule-based dimerization system and demonstrate a sufficiently fast switch-off to determine kinetics of lipid metabolizing enzymes in living cells. We applied this new method to induce and stop phosphatidylinositol 3-kinase (PI3K) activity, allowing us to quantitatively measure the turnover of phosphatidylinositol 3,4,5-trisphosphate (PIP3) and its downstream effectors by confocal fluorescence microscopy as well as standard biochemical methods.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Dimerization , HeLa Cells , Humans , Kinetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tacrolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...