Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Mol Cell ; 71(6): 1092-1104.e5, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30174291

ABSTRACT

Activation of class I phosphatidylinositol 3-kinase (PI3K) leads to formation of phosphatidylinositol-3,4,5-trisphophate (PIP3) and phosphatidylinositol-3,4-bisphophate (PI34P2), which spatiotemporally coordinate and regulate a myriad of cellular processes. By simultaneous quantitative imaging of PIP3 and PI34P2 in live cells, we here show that they have a distinctively different spatiotemporal distribution and history in response to growth factor stimulation, which allows them to selectively induce the membrane recruitment and activation of Akt isoforms. PI34P2 selectively activates Akt2 at both the plasma membrane and early endosomes, whereas PIP3 selectively stimulates Akt1 and Akt3 exclusively at the plasma membrane. These spatiotemporally distinct activation patterns of Akt isoforms provide a mechanism for their differential regulation of downstream signaling molecules. Collectively, our studies show that different spatiotemporal dynamics of PIP3 and PI34P2 and their ability to selectively activate key signaling proteins allow them to mediate class I PI3K signaling pathways in a spatiotemporally specific manner.


Subject(s)
Optical Imaging/methods , Phosphatidylinositol Phosphates/physiology , Single Molecule Imaging/methods , Animals , Cell Line , Cell Membrane , Humans , Inositol Phosphates , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositols , Protein Isoforms , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
2.
Nat Chem Biol ; 19(2): 239-250, 2023 02.
Article in English | MEDLINE | ID: mdl-36229686

ABSTRACT

Membrane lipids control the cellular activity of kinases containing the Src homology 2 (SH2) domain through direct lipid-SH2 domain interactions. Here we report development of new nonlipidic small molecule inhibitors of the lipid-SH2 domain interaction that block the cellular activity of their host proteins. As a pilot study, we evaluated the efficacy of lipid-SH2 domain interaction inhibitors for spleen tyrosine kinase (Syk), which is implicated in hematopoietic malignancies, including acute myeloid leukemia (AML). An optimized inhibitor (WC36) specifically and potently suppressed oncogenic activities of Syk in AML cell lines and patient-derived AML cells. Unlike ATP-competitive Syk inhibitors, WC36 was refractory to de novo and acquired drug resistance due to its ability to block not only the Syk kinase activity, but also its noncatalytic scaffolding function that is linked to drug resistance. Collectively, our study shows that targeting lipid-protein interaction is a powerful approach to developing new small molecule drugs.


Subject(s)
Leukemia, Myeloid, Acute , Protein-Tyrosine Kinases , Humans , Protein-Tyrosine Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Pilot Projects , src Homology Domains , Phosphorylation , Leukemia, Myeloid, Acute/drug therapy , Lipids , Syk Kinase/metabolism
3.
Mol Cell ; 62(1): 7-20, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27052731

ABSTRACT

The Src-homology 2 (SH2) domain is a protein interaction domain that directs myriad phosphotyrosine (pY)-signaling pathways. Genome-wide screening of human SH2 domains reveals that ∼90% of SH2 domains bind plasma membrane lipids and many have high phosphoinositide specificity. They bind lipids using surface cationic patches separate from pY-binding pockets, thus binding lipids and the pY motif independently. The patches form grooves for specific lipid headgroup recognition or flat surfaces for non-specific membrane binding and both types of interaction are important for cellular function and regulation of SH2 domain-containing proteins. Cellular studies with ZAP70 showed that multiple lipids bind its C-terminal SH2 domain in a spatiotemporally specific manner and thereby exert exquisite spatiotemporal control over its protein binding and signaling activities in T cells. Collectively, this study reveals how lipids control SH2 domain-mediated cellular protein-protein interaction networks and suggest a new strategy for therapeutic modulation of pY-signaling pathways.


Subject(s)
Lipid Metabolism , T-Lymphocytes/metabolism , ZAP-70 Protein-Tyrosine Kinase/chemistry , ZAP-70 Protein-Tyrosine Kinase/metabolism , src Homology Domains , Binding Sites , Cells, Cultured , Humans , Jurkat Cells , Models, Molecular , Molecular Docking Simulation , Phosphotyrosine/drug effects , Phosphotyrosine/metabolism , Protein Binding , Protein Structure, Tertiary , Signal Transduction
4.
J Lipid Res ; 63(3): 100178, 2022 03.
Article in English | MEDLINE | ID: mdl-35143844

ABSTRACT

Cholesterol is an essential component of mammalian cell membranes whose subcellular concentration and function are tightly regulated by de novo biosynthesis, transport, and storage. Although recent reports have suggested diverse functions of cellular cholesterol in different subcellular membranes, systematic investigation of its site-specific roles has been hampered by the lack of a methodology for spatiotemporal manipulation of cellular cholesterol levels. Here, we report the development of a new cholesterol depletion system that allows for spatiotemporal manipulation of intracellular cholesterol levels. This system utilizes a genetically encoded cholesterol oxidase whose intrinsic membrane binding activity is engineered in such a way that its membrane targeting can be controlled in a spatiotemporally specific manner via chemically induced dimerization. In combination with in situ quantitative imaging of cholesterol and signaling activity measurements, this system allows for unambiguous determination of site-specific functions of cholesterol in different membranes, including the plasma membrane and the lysosomal membrane.


Subject(s)
Cholesterol , Lysosomes , Animals , Cell Membrane/metabolism , Cholesterol/metabolism , Endosomes/metabolism , Intracellular Membranes/metabolism , Lysosomes/metabolism , Mammals/metabolism
5.
J Lipid Res ; 61(6): 945-952, 2020 06.
Article in English | MEDLINE | ID: mdl-32341006

ABSTRACT

Lipid kinases and phosphatases play key roles in cell signaling and regulation, are implicated in many human diseases, and are thus attractive targets for drug development. Currently, no direct in vitro activity assay is available for these important enzymes, which hampers mechanistic studies as well as high-throughput screening of small molecule modulators. Here, we report a highly sensitive and quantitative assay employing a ratiometric fluorescence sensor that directly and specifically monitors the real-time concentration change of a single lipid species. Because of its modular design, the assay system can be applied to a wide variety of lipid kinases and phosphatases, including class I phosphoinositide 3-kinase (PI3K) and phosphatase and tensin homolog (PTEN). When applied to PI3K, the assay provided detailed mechanistic information about the product inhibition and substrate acyl-chain selectivity of PI3K and enabled rapid evaluation of small molecule inhibitors. We also used this assay to quantitatively determine the substrate specificity of PTEN, providing new insight into its physiological function. In summary, we have developed a fluorescence-based real-time assay for PI3K and PTEN that we anticipate could be adapted to measure the activities of other lipid kinases and phosphatases with high sensitivity and accuracy.


Subject(s)
Enzyme Assays/methods , Fluorometry , Phosphoric Monoester Hydrolases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Substrate Specificity
6.
Plant Cell ; 29(6): 1388-1405, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28584166

ABSTRACT

During cytokinesis in plants, trans-Golgi network-derived vesicles accumulate at the center of dividing cells and undergo various structural changes to give rise to the planar cell plate. However, how this conversion occurs at the molecular level remains elusive. In this study, we report that SH3 Domain-Containing Protein 2 (SH3P2) in Arabidopsis thaliana plays a crucial role in converting vesicles to the planar cell plate. SH3P2 RNAi plants showed cytokinesis-defective phenotypes and produced aggregations of vesicles at the leading edge of the cell plate. SH3P2 localized to the leading edge of the cell plate, particularly the constricted or curved regions of the cell plate. The BAR domain of SH3P2 induced tubulation of vesicles. SH3P2 formed a complex with dynamin-related protein 1A (DRP1A) and affected DRP1A accumulation to the cell plate. Based on these results, we propose that SH3P2 functions together with DRP1A to convert the fused vesicles to tubular structures during cytokinesis.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/cytology , Arabidopsis/metabolism , Carrier Proteins/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Carrier Proteins/genetics , Cytokinesis/genetics , Cytokinesis/physiology , Dynamins/genetics , Dynamins/metabolism , Plants, Genetically Modified/cytology , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , trans-Golgi Network/metabolism , trans-Golgi Network/physiology
7.
Biochemistry ; 56(40): 5318-5327, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28915027

ABSTRACT

In vitro studies of protein structure, function, and dynamics typically preclude the complex range of molecular interactions found in living tissues. In vivo studies elucidate these complex relationships, yet they are typically incompatible with the extensive and controlled biophysical experiments available in vitro. We present an alternative approach by extracting membranes from eukaryotic tissues to produce native bicelles to capture the rich and complex molecular environment of in vivo studies while retaining the advantages of in vitro experiments. Native bicelles derived from chicken egg or mouse cerebrum tissues contain a rich composition of phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylglycerol (PG), phosphatidylserine (PS), phosphatidylinositol (PI), phosphatidic acid (PA), lysolipids, cholesterol, ceramides (CM), and sphingomyelin (SM). The bicelles also contain source-specific lipids such as triacylglycerides (TAGs) and sulfatides from egg and brain tissues, respectively. With the influenza hemagglutinin fusion peptide (HAfp) and the C-terminal Src homology domain of lymphocyte-specific protein-tyrosine kinase (lck-cSH2), we show that membrane proteins and membrane associated proteins reconstituted in native bicelles produce high-resolution NMR data and probe native protein-lipid interactions.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Micelles , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Mice , Models, Molecular , Protein Conformation
8.
J Biol Chem ; 291(34): 17639-50, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27334919

ABSTRACT

Lymphocyte-specific protein-tyrosine kinase (Lck) plays an essential role in T cell receptor (TCR) signaling and T cell development, but its activation mechanism is not fully understood. To explore the possibility that plasma membrane (PM) lipids control TCR signaling activities of Lck, we measured the membrane binding properties of its regulatory Src homology 2 (SH2) and Src homology 3 domains. The Lck SH2 domain binds anionic PM lipids with high affinity but with low specificity. Electrostatic potential calculation, NMR analysis, and mutational studies identified the lipid-binding site of the Lck SH2 domain that includes surface-exposed basic, aromatic, and hydrophobic residues but not the phospho-Tyr binding pocket. Mutation of lipid binding residues greatly reduced the interaction of Lck with the ζ chain in the activated TCR signaling complex and its overall TCR signaling activities. These results suggest that PM lipids, including phosphatidylinositol 4,5-bisphosphate and phosphatidylinositol 3,4,5-trisphosphate, modulate interaction of Lck with its binding partners in the TCR signaling complex and its TCR signaling activities in a spatiotemporally specific manner via its SH2 domain.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/physiology , Amino Acid Substitution , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mutation, Missense , Phosphatidylinositol 4,5-Diphosphate/genetics , Phosphatidylinositol Phosphates/genetics , Receptors, Antigen, T-Cell/genetics , src Homology Domains
9.
bioRxiv ; 2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36747849

ABSTRACT

3'-Phosphoinositides are ubiquitous cellular lipids that play pivotal regulatory roles in health and disease. Generation of 3'-phosphoinositides are driven by three families of phosphoinositide 3-kinases (PI3K) but the mechanisms underlying their regulation and cross-talk are not fully understood. Among 3'-phosphoinositides, phosphatidylinositol-3,5-bisphosphate (PI(3,5)P 2 ) remains the least understood species in terms of its spatiotemporal dynamics and physiological function due to the lack of specific probes. By means of spatiotemporally resolved in situ quantitative imaging of PI(3,5)P 2 using a newly developed ratiometric PI(3,5)P 2 sensor we demonstrate that a special pool of PI(3,5)P 2 is generated on lysosomes and late endosomes in response to growth factor stimulation. This PI(3,5)P 2 pool, the formation of which is mediated by Class II PI3KC2ß and PIKFyve, plays a crucial role in terminating the activity of growth factor-stimulated Class I PI3K, one of the most frequently mutated proteins in cancer, via specific interaction with its regulatory p85 subunit. Cancer-causing mutations of Class I PI3K inhibit the p85-PI(3,5)P 2 interaction and thereby induce sustained activation of Class I PI3K. Our results unravel a hitherto unknown tight regulatory interplay between Class I and II PI3Ks mediated by PI(3,5)P 2 , which may be important for controlling the strength of PI3K-mediated growth factor signaling. These results also suggest a new therapeutic possibility of treating cancer patients with p85 mutations.

10.
ACS Chem Biol ; 15(7): 1913-1920, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32525312

ABSTRACT

Ratiometric fluorescence sensors are powerful tools for direct quantification of diverse biological analytes. To overcome a shortage of solvatochromic fluorophores crucial for in situ ratiometric imaging of biological targets, we prepared and characterized a small library of modular fluorophores with diverse spectral properties. Among them, WCB and WCR showed excellent spectral properties, including high photostability, brightness, and solvatochromism, and are ideally suited for dual ratiometric imaging due to their spectral orthogonality. By conjugating WCB and WCR with protein-based lipid sensors, we were able to achieve robust simultaneous in situ quantitative imaging of two metabolically linked signaling lipids, phosphatidylinositol-4,5-bisphosphate and phosphatidylinositol-3,4,5-trisphosphate in live cells. This study shows that any combination of signaling molecules can be simultaneously quantified in a spatiotemporally resolved manner by ratiometric imaging with finely tuned solvatochromic fluorophores.


Subject(s)
Fluorenes/chemistry , Fluorescent Dyes/chemistry , Phosphatidylinositol Phosphates/analysis , Animals , Fluorenes/chemical synthesis , Fluorescent Dyes/chemical synthesis , Mice , NIH 3T3 Cells , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Spectrometry, Fluorescence
11.
Cell Signal ; 51: 130-138, 2018 11.
Article in English | MEDLINE | ID: mdl-30092354

ABSTRACT

Regulation of tyrosine phosphorylation on insulin receptor substrate-1 (IRS-1) is essential for insulin signaling. The protein tyrosine phosphatase (PTP) C1-Ten/Tensin2 has been implicated in the regulation of IRS-1, but the molecular basis of this dephosphorylation is not fully understood. Here, we demonstrate that the cellular phosphatase activity of C1-Ten/Tensin2 on IRS-1 is mediated by the binding of the C1-Ten/Tensin2 Src-homology 2 (SH2) domain to phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P3). We show that the role of C1-Ten/Tensin2 is dependent on insulin-induced phosphoinositide 3-kinase activity. The C1-Ten/Tensin2 SH2 domain showed strong preference and high affinity for PtdIns(3,4,5)P3. Using site-directed mutagenesis, we identified three basic residues in the C1-Ten/Tensin2 SH2 domain that were critical for PtdIns(3,4,5)P3 binding but were not involved in phosphotyrosine binding and PTP activity. Using a PtdIns(3,4,5)P3 binding-deficient mutant, we showed that the specific binding of the C1-Ten/Tensin2 SH2 domain to PtdIns(3,4,5)P3 allowed C1-Ten/Tensin2 to function as a PTP in cells. Collectively, our findings suggest that the interaction between the C1-Ten/Tensin2 SH2 domain and PtdIns(3,4,5)P3 produces a negative feedback loop of insulin signaling through IRS-1.


Subject(s)
Insulin Receptor Substrate Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Tensins/chemistry , Tensins/metabolism , src Homology Domains , Animals , Escherichia coli , HEK293 Cells , Humans , L Cells , Mice , Mutagenesis, Site-Directed , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Tensins/genetics
12.
J Cell Biol ; 216(11): 3767-3783, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28874417

ABSTRACT

A unique feature of α-catenin localized outside the cadherin-catenin complex is its capacity to form homodimers, but the subcellular localization and functions of this form of α-catenin remain incompletely understood. We identified a cadherin-free form of α-catenin that is recruited to the leading edge of migrating cells in a phosphatidylinositol 3-kinase-dependent manner. Surface plasmon resonance analysis shows that α-catenin homodimers, but not monomers, selectively bind phosphatidylinositol-3,4,5-trisphosphate-containing lipid vesicles with high affinity, where three basic residues, K488, K493, and R496, contribute to binding. Chemical-induced dimerization of α-catenin containing a synthetic dimerization domain promotes its accumulation within lamellipodia and elaboration of protrusions with extended filopodia, which are attenuated in the α-cateninKKR<3A mutant. Cells restored with a full-length, natively homodimerizing form of α-cateninKKR<3A display reduced membrane recruitment, altered epithelial sheet migrations, and weaker cell-cell adhesion compared with WT α-catenin. These findings show that α-catenin homodimers are recruited to phosphoinositide-activated membranes to promote adhesion and migration, suggesting that phosphoinositide binding may be a defining feature of α-catenin function outside the cadherin-catenin complex.


Subject(s)
Cell Adhesion , Cell Membrane/metabolism , Epithelial Cells/metabolism , Phosphatidylinositol Phosphates/metabolism , alpha Catenin/metabolism , Animals , Cell Line, Tumor , Cell Movement , Dogs , Humans , Madin Darby Canine Kidney Cells , Mutation , Phosphatidylinositol 3-Kinase/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Pseudopodia/metabolism , Signal Transduction , Time Factors , Transfection , alpha Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL