Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 328
Filter
1.
Nat Commun ; 15(1): 4986, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862544

ABSTRACT

Focal adhesions form liquid-like assemblies around activated integrin receptors at the plasma membrane. How they achieve their flexible properties is not well understood. Here, we use recombinant focal adhesion proteins to reconstitute the core structural machinery in vitro. We observe liquid-liquid phase separation of the core focal adhesion proteins talin and vinculin for a spectrum of conditions and interaction partners. Intriguingly, we show that binding to PI(4,5)P2-containing membranes triggers phase separation of these proteins on the membrane surface, which in turn induces the enrichment of integrin in the clusters. We suggest a mechanism by which 2-dimensional biomolecular condensates assemble on membranes from soluble proteins in the cytoplasm: lipid-binding triggers protein activation and thus, liquid-liquid phase separation of these membrane-bound proteins. This could explain how early focal adhesions maintain a structured and force-resistant organization into the cytoplasm, while still being highly dynamic and able to quickly assemble and disassemble.


Subject(s)
Cell Membrane , Focal Adhesions , Talin , Vinculin , Talin/metabolism , Talin/chemistry , Focal Adhesions/metabolism , Cell Membrane/metabolism , Vinculin/metabolism , Vinculin/chemistry , Humans , Animals , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Integrins/metabolism , Integrins/chemistry , Cytoplasm/metabolism , Protein Binding , Phase Separation
2.
Biophys J ; 123(16): 2431-2442, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38859585

ABSTRACT

Annexin A2 (A2)-induced microdomain formation is a key step in biological processes such as Ca2+-mediated exocytosis in neuroendocrine cells. In this work, a total of 15 coarse-grained molecular dynamics simulations were performed on vesicle models having a diameter of approximately 250 Å for 15 µs each using the Martini2 force field. Five simulations were performed in the presence of 10 A2, 5 in the presence of A2 but absence of PIP2, and 5 simulations in the absence of A2 but presence of PIP2. Consistent results were generated among the simulations. A2-induced PIP2 microdomain formation was observed and shown to occur in three phases: A2-vesicle association, localized A2-induced PIP2 clustering, and A2 aggregation driving PIP2 microdomain formation. The relationship between A2 aggregation and PIP2 microdomain formation was quantitatively described using a novel method which calculated the variance among protein and lipid positions via the Fréchet mean. A large reduction in PIP2 variance was observed in the presence of A2 but not in its absence. This reduction in PIP2 variance was proportional to the reduction observed in A2 variance and demonstrates that the observed PIP2 microdomain formation is dependent upon A2 aggregation. The three-phase model of A2-induced microdomain formation generated in this work will serve as a valuable guide for further experimental studies and the development of novel A2 inhibitors. No microdomain formation was observed in the absence of A2 and minimal A2-membrane interaction was observed in the absence of PIP2.


Subject(s)
Annexin A2 , Molecular Dynamics Simulation , Annexin A2/metabolism , Annexin A2/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Membrane Microdomains/metabolism , Membrane Microdomains/chemistry
3.
ACS Nano ; 18(20): 12737-12748, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38717305

ABSTRACT

Lipids are key factors in regulating membrane fusion. Lipids are not only structural components to form membranes but also active catalysts for vesicle fusion and neurotransmitter release, which are driven by soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins. SNARE proteins seem to be partially assembled before fusion, but the mechanisms that arrest vesicle fusion before Ca2+ influx are still not clear. Here, we show that phosphatidylinositol 4,5-bisphosphate (PIP2) electrostatically triggers vesicle fusion as an electrostatic catalyst by lowering the hydration energy and that a myristoylated alanine-rich C-kinase substrate (MARCKS), a PIP2-binding protein, arrests vesicle fusion in a vesicle docking state where the SNARE complex is partially assembled. Vesicle-mimicking liposomes fail to reproduce vesicle fusion arrest by masking PIP2, indicating that native vesicles are essential for the reconstitution of physiological vesicle fusion. PIP2 attracts cations to repel water molecules from membranes, thus lowering the hydration energy barrier.


Subject(s)
Membrane Fusion , Phosphatidylinositol 4,5-Diphosphate , Static Electricity , Water , Catalysis , Liposomes/chemistry , Membrane Fusion/drug effects , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/pharmacology , SNARE Proteins/metabolism , SNARE Proteins/chemistry , Water/chemistry
4.
FEBS Lett ; 598(11): 1402-1410, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38589226

ABSTRACT

Overactivation of the epidermal growth factor receptor (EGFR) is critical for the development of multiple cancers. Previous studies have shown that the cell membrane is a key regulator of EGFR kinase activity through its interaction with the EGFR juxtamembrane domain (JM). However, the lipid recognition specificity of EGFR-JM and its interaction details remain unclear. Using lipid strip and liposome pulldown assays, we showed that EGFR-JM could specifically interact with PI(4,5)P2-or phosphatidylserine-containing membranes. We further characterized the JM-membrane interaction using NMR-titration-based chemical shift perturbation and paramagnetic relaxation enhancement analyses, and found that residues I649 - L659 comprised the membrane-binding site. Furthermore, the membrane-binding region contains the predicted dimerization motif of JM, 655LRRLL659, suggesting that membrane binding may affect JM dimerization and, therefore, regulate kinase activation.


Subject(s)
Cell Membrane , ErbB Receptors , Phosphatidylserines , Protein Binding , Protein Domains , ErbB Receptors/metabolism , ErbB Receptors/chemistry , ErbB Receptors/genetics , Humans , Cell Membrane/metabolism , Phosphatidylserines/metabolism , Phosphatidylserines/chemistry , Binding Sites , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Liposomes/metabolism , Liposomes/chemistry , Protein Multimerization , Amino Acid Sequence
5.
J Phys Chem B ; 128(9): 2134-2143, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38393820

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PIP2) is a critical lipid for cellular signaling. The specific phosphorylation of the inositol ring controls protein binding as well as clustering behavior. Two popular models to describe ion-mediated clustering of PIP2 are Martini3 (M3) and CHARMM36 (C36). Molecular dynamics simulations of PIP2-containing bilayers in solutions of potassium chloride, sodium chloride, and calcium chloride, and at two different resolutions are performed to understand the aggregation and the model parameters that drive it. The average M3 clusters of PIP2 in bilayers of 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine and PIP2 bilayers in the presence of K+, Na+, or Ca2+ contained 2.2, 2.6, and 6.4 times more PIP2 than C36 clusters, respectively. Indeed, the Ca2+-containing systems often formed a single large aggregate. Reparametrization of the M3 ion-phosphate Lennard-Jones interaction energies to reproduce experimental osmotic pressure of sodium dimethyl phosphate (DMP), K[DMP], and Ca[DMP]2 solutions, the same experimental target as C36, yielded comparably sized PIP2 clusters for the two models. Furthermore, C36 and the modified M3 predict similar saturation of the phosphate groups with increasing Ca2+, although the coarse-grained model does not capture the cooperativity between K+ and Ca2+. This characterization of the M3 behavior in the presence of monovalent and divalent ions lays a foundation to study cation/protein/PIP2 clustering.


Subject(s)
Molecular Dynamics Simulation , Phosphatidylinositol 4,5-Diphosphate , Phosphatidylinositol 4,5-Diphosphate/chemistry , Cations , Sodium
6.
Biophys J ; 123(14): 2001-2011, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38142298

ABSTRACT

Cell signaling is an important process involving complex interactions between lipids and proteins. The myristoylated alanine-rich C-kinase substrate (MARCKS) has been established as a key signaling regulator, serving a range of biological roles. Its effector domain (ED), which anchors the protein to the plasma membrane, induces domain formation in membranes containing phosphatidylinositol 4,5-bisphosphate (PIP2) and phosphatidylserine (PS). The mechanisms governing the MARCKS-ED binding to membranes remain elusive. Here, we investigate the composition-dependent affinity and MARCKS-ED-binding-induced changes in interfacial environments using two-dimensional infrared spectroscopy and fluorescence anisotropy. Both negatively charged lipids facilitate the MARCKS-ED binding to lipid vesicles. Although the hydrogen-bonding structure at the lipid-water interface remains comparable across vesicles with varied lipid compositions, the dynamics of interfacial water show divergent patterns due to specific interactions between lipids and peptides. Our findings also reveal that PIP2 becomes sequestered by bound peptides, while the distribution of PS exhibits no discernible change upon peptide binding. Interestingly, PIP2 and PS become colocalized into domains both in the presence and absence of MARCKS-ED. More broadly, this work offers molecular insights into the effects of membrane composition on binding.


Subject(s)
Myristoylated Alanine-Rich C Kinase Substrate , Phosphatidylinositol 4,5-Diphosphate , Phosphatidylserines , Protein Binding , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Myristoylated Alanine-Rich C Kinase Substrate/chemistry , Peptides/chemistry , Peptides/metabolism , Protein Domains , Water/chemistry , Cell Membrane/metabolism , Cell Membrane/chemistry , Amino Acid Sequence
7.
J Mol Biol ; 435(17): 168193, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37406927

ABSTRACT

Dysferlin is a large membrane protein found most prominently in striated muscle. Loss of dysferlin activity is associated with reduced exocytosis, abnormal intracellular Ca2+ and the muscle diseases limb-girdle muscular dystrophy and Miyoshi myopathy. The cytosolic region of dysferlin consists of seven C2 domains with mutations in the C2A domain at the N-terminus resulting in pathology. Despite the importance of Ca2+ and membrane binding activities of the C2A domain for dysferlin function, the mechanism of the domain remains poorly characterized. In this study we find that the C2A domain preferentially binds membranes containing PI(4,5)P2 through an interaction mediated by residues Y23, K32, K33, and R77 on the concave face of the domain. We also found that subsequent to membrane binding, the C2A domain inserts residues on the Ca2+ binding loops into the membrane. Analysis of solution NMR measurements indicate that the domain inhabits two distinct structural states, with Ca2+ shifting the population between states towards a more rigid structure with greater affinity for PI(4,5)P2. Based on our results, we propose a mechanism where Ca2+ converts C2A from a structurally dynamic, low PI(4,5)P2 affinity state to a high affinity state that targets dysferlin to PI(4,5)P2 enriched membranes through interaction with Tyr23, K32, K33, and R77. Binding also involves changes in lipid packing and insertion by the third Ca2+ binding loop of the C2 domain into the membrane, which would contribute to dysferlin function in exocytosis and Ca2+ regulation.


Subject(s)
Calcium-Binding Proteins , Calcium , Dysferlin , Membrane Proteins , Phosphatidylinositol 4,5-Diphosphate , Calcium/metabolism , Calcium-Binding Proteins/chemistry , Dysferlin/chemistry , Dysferlin/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , C2 Domains , Protein Binding , Phosphatidylinositol 4,5-Diphosphate/chemistry
8.
Langmuir ; 39(17): 5995-6005, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37086192

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PIP2) is an important molecule located at the inner leaflet of cell membrane, where it serves as anchoring sites for a cohort of membrane-associated molecules and as a broad-reaching signaling intermediate. The lipid raft is thought as the major platform recruiting proteins for signal transduction and also known to mediate PIP2 accumulation across the membrane. While the significance of this cross-membrane coupling is increasingly appreciated, it remains unclear whether and how PIP2 senses the dynamic change of the ordered lipid domains over the packed hydrophobic core of the bilayer. Herein, by means of molecular dynamic simulation, we reveal that inner PIP2 molecules can sense the outer lipid domain via inter-leaflet coupling, and the coupling manner is dictated by the acyl chain length of sphingomyelin (SM) partitioned to the lipid domain. Shorter SM promotes membrane domain registration, whereby PIP2 accumulates beneath the domain across the membrane. In contrast, the anti-registration is thermodynamically preferred if the lipid domain has longer SM due to the hydrophobic mismatch between the corresponding acyl chains in SM and PIP2. In this case, PIP2 is expelled by the domain with a higher diffusivity. These results provide molecular insights into the regulatory mechanism of correlation between the outer lipid domain and inner PIP2, both of which are critical components for cell signal transduction.


Subject(s)
Phosphatidylinositols , Sphingomyelins , Humans , Phosphatidylinositols/analysis , Phosphatidylinositols/metabolism , Cell Membrane/chemistry , Molecular Dynamics Simulation , Membrane Microdomains/chemistry , Phosphatidylinositol 4,5-Diphosphate/analysis , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism
9.
Adv Exp Med Biol ; 1422: 61-85, 2023.
Article in English | MEDLINE | ID: mdl-36988877

ABSTRACT

Cell membranes regulate a wide range of phenomena that are implicated in key cellular functions. Cholesterol, a critical component of eukaryotic cell membranes, is responsible for cellular organization, membrane elasticity, and other critical physicochemical parameters. Besides cholesterol, other lipid components such as phosphatidylinositol 4,5-bisphosphate (PIP2) are found in minor concentrations in cell membranes yet can also play a major regulatory role in various cell functions. In this chapter, we describe how solid-state deuterium nuclear magnetic resonance (2H NMR) spectroscopy together with neutron spin-echo (NSE) spectroscopy can inform synergetic changes to lipid molecular packing due to cholesterol and PIP2 that modulate the bending rigidity of lipid membranes. Fundamental structure-property relations of molecular self-assembly are illuminated and point toward a length and time-scale dependence of cell membrane mechanics, with significant implications for biological activity and membrane lipid-protein interactions.


Subject(s)
Membrane Lipids , Phosphatidylinositols , Phosphatidylinositols/metabolism , Cell Membrane/metabolism , Membrane Lipids/metabolism , Cholesterol/chemistry , Biophysics , Lipid Bilayers/chemistry , Phosphatidylinositol 4,5-Diphosphate/analysis , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism
10.
Adv Exp Med Biol ; 1422: 381-392, 2023.
Article in English | MEDLINE | ID: mdl-36988889

ABSTRACT

Besides its protective role in the maintenance of cell homeostasis, the plasma membrane is the site of exchanges between the cell interior and the extracellular medium. To circumvent the hydrophobic barrier formed by the acyl chains of the lipid bilayer, protein channels and transporters are key players in the exchange of small hydrophilic compounds such as ions or nutrients, but they hardly account for the transport of larger biological molecules. Exchange of proteins usually relies on membrane-fusion events between vesicles and the plasma membrane. In recent years, several alternative unconventional protein secretion (UPS) pathways across the plasma membrane have been characterised for a specific set of secreted substrates, some of them excluding any membrane-fusion events (Dimou and Nickel, Curr Biol 28:R406-R410, 2018). One of thesbe pathways, referred as type I UPS, relies on the direct translocation of the protein across the plasma membrane and not surprisingly, lipids are essential players in this process. In this chapter, we discuss the roles of phosphatidylinositol(4,5)bisphosphate (PI(4,5)P2) and cholesterol in unconventional pathways involving Engrailed-2 homeoprotein and fibroblast growth factor 2.


Subject(s)
Lipid Bilayers , Membrane Transport Proteins , Lipid Bilayers/metabolism , Cell Membrane/metabolism , Protein Transport , Membrane Transport Proteins/metabolism , Cholesterol/metabolism , Phosphatidylinositol 4,5-Diphosphate/analysis , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism
11.
Biol Chem ; 404(4): 241-254, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36809224

ABSTRACT

The Phosphatidylinositol 3-phosphate 5-kinase Type III PIKfyve is the main source for selectively generated phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), a known regulator of membrane protein trafficking. PI(3,5)P2 facilitates the cardiac KCNQ1/KCNE1 channel plasma membrane abundance and therewith increases the macroscopic current amplitude. Functional-physical interaction of PI(3,5)P2 with membrane proteins and its structural impact is not sufficiently understood. This study aimed to identify molecular interaction sites and stimulatory mechanisms of the KCNQ1/KCNE1 channel via the PIKfyve-PI(3,5)P2 axis. Mutational scanning at the intracellular membrane leaflet and nuclear magnetic resonance (NMR) spectroscopy identified two PI(3,5)P2 binding sites, the known PIP2 site PS1 and the newly identified N-terminal α-helix S0 as relevant for functional PIKfyve effects. Cd2+ coordination to engineered cysteines and molecular modeling suggest that repositioning of S0 stabilizes the channel s open state, an effect strictly dependent on parallel binding of PI(3,5)P2 to both sites.


Subject(s)
KCNQ1 Potassium Channel , Phosphatidylinositol 4,5-Diphosphate , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , KCNQ1 Potassium Channel/chemistry , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Binding Sites , Mutation , Cell Membrane/metabolism
12.
J Colloid Interface Sci ; 629(Pt B): 785-795, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36195018

ABSTRACT

HYPOTHESIS: Inositol phospholipids are well known to form clusters in the cytoplasmic leaflet of the plasma membrane that are responsible for the interaction and recruitment of proteins involved in key biological processes like endocytosis, ion channel activation and secondary messenger production. Although their phosphorylated inositol ring headgroup plays an important role in protein binding, its orientation with respect to the plane of the membrane and its lateral packing density has not been previously described experimentally. EXPERIMENTS: Here, we study phosphatidylinositol 4,5-bisphosphate (PIP2) planar model membranes in the form of Langmuir monolayers by surface pressure-area isotherms, Brewster angle microscopy and neutron reflectometry to elucidate the relation between lateral (in-plane) and perpendicular (out-of-plane) molecular organization of PIP2. FINDINGS: Different surface areas were explored through monolayer compression, allowing us to correlate the formation of transient PIP2 clusters with the change in orientation of the inositol-biphosphate headgroup, which was experimentally determined by neutron reflectometry.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate , Phosphatidylinositols , Phosphatidylinositols/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Inositol Phosphates/metabolism , Cell Membrane/metabolism , Protein Binding
13.
Science ; 378(6616): eadd1268, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36227998

ABSTRACT

The transient receptor potential melastatin 8 (TRPM8) channel is the primary molecular transducer responsible for the cool sensation elicited by menthol and cold in mammals. TRPM8 activation is controlled by cooling compounds together with the membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2). Our knowledge of cold sensation and the therapeutic potential of TRPM8 for neuroinflammatory diseases and pain will be enhanced by understanding the structural basis of cooling agonist- and PIP2-dependent TRPM8 activation. We present cryo-electron microscopy structures of mouse TRPM8 in closed, intermediate, and open states along the ligand- and PIP2-dependent gating pathway. Our results uncover two discrete agonist sites, state-dependent rearrangements in the gate positions, and a disordered-to-ordered transition of the gate-forming S6-elucidating the molecular basis of chemically induced cool sensation in mammals.


Subject(s)
Cold Temperature , Ion Channel Gating , Phosphatidylinositol 4,5-Diphosphate , Pyrimidinones , TRPM Cation Channels , Thermosensing , Animals , Mice , Cryoelectron Microscopy , Ligands , Menthol/chemistry , Menthol/pharmacology , TRPM Cation Channels/agonists , TRPM Cation Channels/chemistry , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Thermosensing/drug effects , Thermosensing/physiology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Protein Conformation , Pyrimidinones/chemistry , Pyrimidinones/pharmacology
14.
Proc Natl Acad Sci U S A ; 119(38): e2208337119, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36103579

ABSTRACT

Synchronous release at neuronal synapses is accomplished by a machinery that senses calcium influx and fuses the synaptic vesicle and plasma membranes to release neurotransmitters. Previous studies suggested the calcium sensor synaptotagmin (Syt) is a facilitator of vesicle docking and both a facilitator and inhibitor of fusion. On phospholipid monolayers, the Syt C2AB domain spontaneously oligomerized into rings that are disassembled by Ca2+, suggesting Syt rings may clamp fusion as membrane-separating "washers" until Ca2+-mediated disassembly triggers fusion and release [J. Wang et al., Proc. Natl. Acad. Sci. U.S.A. 111, 13966-13971 (2014)].). Here, we combined mathematical modeling with experiment to measure the mechanical properties of Syt rings and to test this mechanism. Consistent with experimental results, the model quantitatively recapitulates observed Syt ring-induced dome and volcano shapes on phospholipid monolayers and predicts rings are stabilized by anionic phospholipid bilayers or bulk solution with ATP. The selected ring conformation is highly sensitive to membrane composition and bulk ATP levels, a property that may regulate vesicle docking and fusion in ATP-rich synaptic terminals. We find the Syt molecules hosted by a synaptic vesicle oligomerize into a halo, unbound from the vesicle, but in proximity to sufficiently phosphatidylinositol 4,5-bisphosphate (PIP2)-rich plasma membrane (PM) domains, the PM-bound trans Syt ring conformation is preferred. Thus, the Syt halo serves as landing gear for spatially directed docking at PIP2-rich sites that define the active zones of exocytotic release, positioning the Syt ring to clamp fusion and await calcium. Our results suggest the Syt ring is both a Ca2+-sensitive fusion clamp and a high-fidelity sensor for directed docking.


Subject(s)
Synaptic Vesicles , Synaptotagmin I , Adenosine Triphosphate/metabolism , Calcium/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Synaptic Vesicles/metabolism , Synaptotagmin I/chemistry
15.
J Biol Chem ; 298(8): 102264, 2022 08.
Article in English | MEDLINE | ID: mdl-35843309

ABSTRACT

TransMEMbrane 16A (TMEM16A) is a Ca2+-activated Cl- channel that plays critical roles in regulating diverse physiologic processes, including vascular tone, sensory signal transduction, and mucosal secretion. In addition to Ca2+, TMEM16A activation requires the membrane lipid phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). However, the structural determinants mediating this interaction are not clear. Here, we interrogated the parts of the PI(4,5)P2 head group that mediate its interaction with TMEM16A by using patch- and two-electrode voltage-clamp recordings on oocytes from the African clawed frog Xenopus laevis, which endogenously express TMEM16A channels. During continuous application of Ca2+ to excised inside-out patches, we found that TMEM16A-conducted currents decayed shortly after patch excision. Following this rundown, we show that the application of a synthetic PI(4,5)P2 analog produced current recovery. Furthermore, inducible dephosphorylation of PI(4,5)P2 reduces TMEM16A-conducted currents. Application of PIP2 analogs with different phosphate orientations yielded distinct amounts of current recovery, and only lipids that include a phosphate at the 4' position effectively recovered TMEM16A currents. Taken together, these findings improve our understanding of how PI(4,5)P2 binds to and potentiates TMEM16A channels.


Subject(s)
Phosphates , Phosphatidylinositol 4,5-Diphosphate , Animals , Calcium/metabolism , Chloride Channels/metabolism , Phosphates/chemistry , Phosphates/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Xenopus laevis/metabolism
16.
Proc Natl Acad Sci U S A ; 119(25): e2202295119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35696574

ABSTRACT

Caveolae are small plasma membrane invaginations, important for control of membrane tension, signaling cascades, and lipid sorting. The caveola coat protein Cavin1 is essential for shaping such high curvature membrane structures. Yet, a mechanistic understanding of how Cavin1 assembles at the membrane interface is lacking. Here, we used model membranes combined with biophysical dissection and computational modeling to show that Cavin1 inserts into membranes. We establish that initial phosphatidylinositol (4, 5) bisphosphate [PI(4,5)P2]-dependent membrane adsorption of the trimeric helical region 1 (HR1) of Cavin1 mediates the subsequent partial separation and membrane insertion of the individual helices. Insertion kinetics of HR1 is further enhanced by the presence of flanking negatively charged disordered regions, which was found important for the coassembly of Cavin1 with Caveolin1 in living cells. We propose that this intricate mechanism potentiates membrane curvature generation and facilitates dynamic rounds of assembly and disassembly of Cavin1 at the membrane.


Subject(s)
Caveolae , RNA-Binding Proteins , Caveolae/chemistry , Caveolin 1/chemistry , HEK293 Cells , Humans , Phosphatidylinositol 4,5-Diphosphate/chemistry , Protein Domains , Protein Transport , RNA-Binding Proteins/chemistry , Signal Transduction
17.
Nano Lett ; 22(3): 1449-1455, 2022 02 09.
Article in English | MEDLINE | ID: mdl-34855407

ABSTRACT

A mechanism for full-length synaptotagmin-1 (syt-1) to interact with anionic bilayers and to promote fusion in the presence of SNAREs is proposed. Colloidal probe force spectroscopy in conjunction with tethered particle motion monitoring showed that in the absence of Ca2+ the binding of syt-1 to membranes depends on the presence and content of PI(4,5)P2. Addition of Ca2+ switches the interaction forces from weak to strong, eventually exceeding the cohesion of the C2A domain of syt-1 leading to partial unfolding of the protein. Fusion of single unilamellar vesicles equipped with syt-1 and synaptobrevin 2 with planar pore-spanning target membranes containing PS and PI(4,5)P2 shows an almost complete suppression of stalled intermediate fusion states and an accelerated fusion kinetics in the presence of Ca2+, which is further enhanced upon addition of ATP.


Subject(s)
Calcium , Phosphatidylinositol 4,5-Diphosphate , SNARE Proteins , Synaptotagmin I , Calcium/chemistry , Calcium/metabolism , Kinetics , Membrane Fusion , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Unfolding , Synaptotagmin I/chemistry , Synaptotagmin I/metabolism
18.
Biochim Biophys Acta Biomembr ; 1863(12): 183757, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34478732

ABSTRACT

GLUT1 is a major glucose facilitator expressed ubiquitously among tissues. Upregulation of its expression plays an important role in the development of many types of cancer and metabolic diseases. Thioredoxin-interacting protein (TXNIP) is an α-arrestin that acts as an adaptor for GLUT1 in clathrin-mediated endocytosis. It regulates cellular glucose uptake in response to both intracellular and extracellular signals via its control on GLUT1-4. In order to understand the interaction between GLUT1 and TXNIP, we generated GLUT1 lipid nanodiscs and carried out isothermal titration calorimetry and single-particle electron microscopy experiments. We found that GLUT1 lipid nanodiscs and TXNIP interact in a 1:1 ratio and that this interaction requires phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2 or PIP2).


Subject(s)
Carrier Proteins/genetics , Glucose Transporter Type 1/genetics , Lipids/genetics , Phosphatidylinositol 4,5-Diphosphate/chemistry , Biological Transport/genetics , Carrier Proteins/chemistry , Clathrin/chemistry , Endocytosis/genetics , Glucose/metabolism , Glucose Transporter Type 1/chemistry , Humans , Lipids/chemistry , Phosphatidylinositol 4,5-Diphosphate/genetics , Signal Transduction
19.
Phys Chem Chem Phys ; 23(29): 15784-15795, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34286758

ABSTRACT

G protein-gated inwardly rectifying potassium (GIRK) channels play essential roles in electrical signaling in neurons and muscle cells. Nonequilibrium environments provide crucial driving forces behind many cellular events. Here, we apply the antiparallel alignment double bilayer model to study GIRK2 in response to the time-dependent membrane potential. Using molecular dynamics and umbrella sampling, we examined the time-dependent environmental impact on the ion conduction, energy basis, and primary motions of GIRK2 in different complex states with phosphatidylinositol-4,5-bisphosphate (PIP2) and G-protein ßγ subunits (Gßγ). The antiparallel alignment double bilayer model enables us to study the transport performance in inward and outward K+ and mixed K+ and Na+. We obtained the recoverable discharge process of GIRK2 complexed with both PIP2 and Gßγ, compared with occasional conduction under PIP2-only regulation. Calculations of potential of mean force suggest different regulation by the helix bundle crossing (HBC) gate and G-loop gate regarding different complex states and under a membrane potential. In a nonequilibrium environment, distinct functional rocking motions of GIRK2 were identified under strengthened correlations between the transmembrane helices and downstream cytoplasmic domains with binding of PIP2, cations, and Gßγ. The findings suggest the potential domain motions and dynamics associated with a nonequilibrium environment and highlight the application of the antiparallel alignment double bilayer model to investigate factors in an asymmetric environment.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , Cations/chemistry , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , Membrane Potentials , Molecular Dynamics Simulation , Phosphatidylinositol 4,5-Diphosphate/chemistry , Potassium/chemistry , Protein Conformation , Sodium/chemistry , Thermodynamics
20.
Nat Commun ; 12(1): 3938, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168117

ABSTRACT

The membrane is an integral component of the G protein-coupled receptor signaling machinery. Here we demonstrate that lipids regulate the signaling efficacy and selectivity of the ghrelin receptor GHSR through specific interactions and bulk effects. We find that PIP2 shifts the conformational equilibrium of GHSR away from its inactive state, favoring basal and agonist-induced G protein activation. This occurs because of a preferential binding of PIP2 to specific intracellular sites in the receptor active state. Another lipid, GM3, also binds GHSR and favors G protein activation, but mostly in a ghrelin-dependent manner. Finally, we find that not only selective interactions but also the thickness of the bilayer reshapes the conformational repertoire of GHSR, with direct consequences on G protein selectivity. Taken together, this data illuminates the multifaceted role of the membrane components as allosteric modulators of how ghrelin signal could be propagated.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate/metabolism , Receptors, Ghrelin/chemistry , Receptors, Ghrelin/metabolism , Allosteric Regulation , Binding Sites , Cell Membrane/chemistry , Cell Membrane/metabolism , Cysteine/genetics , Fluorescence Resonance Energy Transfer , G(M3) Ganglioside/metabolism , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Lipid Metabolism , Lipids/chemistry , Mutation , Phosphatidylinositol 4,5-Diphosphate/chemistry , Protein Conformation , Receptors, Ghrelin/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL