Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
1.
J Med Chem ; 67(1): 165-179, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38117948

ABSTRACT

Cytoplasmic vacuolation-associated cell death, known as methuosis, offers a promising nonapoptotic approach for cancer treatment. In this study, we outline the synthesis and evaluation of potent methuosis-inducing compounds. These compounds selectively induce cell death, characterized by extensive cytoplasmic vacuolation in HeLa and MDA-MB-231 cells. Notably, compound L22 exhibited a remarkable interaction with PIKfyve kinase, boasting a Kd value of 0.47 nM, surpassing the positive controls D-13 and MOMIPP in potency. Furthermore, it is important to highlight that cell death induced by compound L22 is unequivocally attributed to methuosis as it differs from apoptosis, necrosis, or autophagy. Importantly, when administered orally, L22 effectively inhibited tumor growth in a HeLa xenograft model without any apparent signs of toxicity. These results underscore the potential of L22 as a valuable tool for in-depth investigations into the mechanisms of methuosis and as a promising lead compound to guide structural optimization.


Subject(s)
Antineoplastic Agents , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Death , Apoptosis , Phosphatidylinositol Phosphates/pharmacology
2.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Article in English | MEDLINE | ID: mdl-35131932

ABSTRACT

Transient receptor potential mucolipin 1 (TRPML1) is a Ca2+-permeable, nonselective cation channel ubiquitously expressed in the endolysosomes of mammalian cells and its loss-of-function mutations are the direct cause of type IV mucolipidosis (MLIV), an autosomal recessive lysosomal storage disease. TRPML1 is a ligand-gated channel that can be activated by phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2] as well as some synthetic small-molecule agonists. Recently, rapamycin has also been shown to directly bind and activate TRPML1. Interestingly, both PI(3,5)P2 and rapamycin have low efficacy in channel activation individually but together they work cooperatively and activate the channel with high potency. To reveal the structural basis underlying the synergistic activation of TRPML1 by PI(3,5)P2 and rapamycin, we determined the high-resolution cryoelectron microscopy (cryo-EM) structures of the mouse TRPML1 channel in various states, including apo closed, PI(3,5)P2-bound closed, and PI(3,5)P2/temsirolimus (a rapamycin analog)-bound open states. These structures, combined with electrophysiology, elucidate the molecular details of ligand binding and provide structural insight into how the TRPML1 channel integrates two distantly bound ligand stimuli and facilitates channel opening.


Subject(s)
Phosphatidylinositol Phosphates/pharmacology , Sirolimus/pharmacology , Transient Receptor Potential Channels/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Transient Receptor Potential Channels/genetics
3.
Structure ; 30(1): 139-155.e5, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34453887

ABSTRACT

Transient receptor potential (TRP) channels emerged in fungi as mechanosensitive osmoregulators. The Saccharomyces cerevisiae vacuolar TRP yeast 1 (TRPY1) is the most studied TRP channel from fungi, but the structure and details of channel modulation remain elusive. Here, we describe the full-length cryoelectron microscopy structure of TRPY1 at 3.1 Å resolution in a closed state. The structure, despite containing an evolutionarily conserved and archetypical transmembrane domain, reveals distinctive structural folds for the cytosolic N and C termini, compared with other eukaryotic TRP channels. We identify an inhibitory phosphatidylinositol 3-phosphate (PI(3)P) lipid-binding site, along with two Ca2+-binding sites: a cytosolic site, implicated in channel activation and a vacuolar lumen site, implicated in inhibition. These findings, together with data from microsecond-long molecular dynamics simulations and a model of a TRPY1 open state, provide insights into the basis of TRPY1 channel modulation by lipids and Ca2+, and the molecular evolution of TRP channels.


Subject(s)
Calcium/metabolism , Phosphatidylinositol Phosphates/pharmacology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism , Binding Sites , Cryoelectron Microscopy , Cytosol/metabolism , Models, Molecular , Molecular Dynamics Simulation , Phosphatidylinositol Phosphates/chemistry , Protein Conformation , Protein Domains , Protein Multimerization , Saccharomyces cerevisiae/chemistry
4.
Nature ; 592(7854): 469-473, 2021 04.
Article in English | MEDLINE | ID: mdl-33762731

ABSTRACT

Serotonin, or 5-hydroxytryptamine (5-HT), is an important neurotransmitter1,2 that activates the largest subtype family of G-protein-coupled receptors3. Drugs that target 5-HT1A, 5-HT1D, 5-HT1E and other 5-HT receptors are used to treat numerous disorders4. 5-HT receptors have high levels of basal activity and are subject to regulation by lipids, but the structural basis for the lipid regulation and basal activation of these receptors and the pan-agonism of 5-HT remains unclear. Here we report five structures of 5-HT receptor-G-protein complexes: 5-HT1A in the apo state, bound to 5-HT or bound to the antipsychotic drug aripiprazole; 5-HT1D bound to 5-HT; and 5-HT1E in complex with a 5-HT1E- and 5-HT1F-selective agonist, BRL-54443. Notably, the phospholipid phosphatidylinositol 4-phosphate is present at the G-protein-5-HT1A interface, and is able to increase 5-HT1A-mediated G-protein activity. The receptor transmembrane domain is surrounded by cholesterol molecules-particularly in the case of 5-HT1A, in which cholesterol molecules are directly involved in shaping the ligand-binding pocket that determines the specificity for aripiprazol. Within the ligand-binding pocket of apo-5-HT1A are structured water molecules that mimic 5-HT to activate the receptor. Together, our results address a long-standing question of how lipids and water molecules regulate G-protein-coupled receptors, reveal how 5-HT acts as a pan-agonist, and identify the determinants of drug recognition in 5-HT receptors.


Subject(s)
Cryoelectron Microscopy , Ligands , Lipids , Receptors, Serotonin, 5-HT1/metabolism , Receptors, Serotonin, 5-HT1/ultrastructure , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Aripiprazole/metabolism , Aripiprazole/pharmacology , Binding Sites , Cholesterol/pharmacology , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/ultrastructure , Humans , Models, Molecular , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol Phosphates/pharmacology , Receptor, Serotonin, 5-HT1A/chemistry , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/ultrastructure , Receptors, Serotonin, 5-HT1/chemistry , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Agonists/metabolism , Serotonin 5-HT1 Receptor Agonists/pharmacology , Water/chemistry
5.
Front Immunol ; 11: 532225, 2020.
Article in English | MEDLINE | ID: mdl-33117337

ABSTRACT

Despite intensive antimicrobial and anti-inflammatory therapies, cystic fibrosis (CF) patients are subjected to chronic infections due to opportunistic pathogens, including multidrug resistant (MDR) Pseudomonas aeruginosa. Macrophages from CF patients show many evidences of reduced phagocytosis in terms of internalization capability, phagosome maturation, and intracellular bacterial killing. In this study, we investigated if apoptotic body-like liposomes (ABLs) loaded with phosphatidylinositol 5-phosphate (PI5P), known to regulate actin dynamics and vesicular trafficking, could restore phagocytic machinery while limiting inflammatory response in in vitro and in vivo models of MDR P. aeruginosa infection. Our results show that the in vitro treatment with ABL carrying PI5P (ABL/PI5P) enhances bacterial uptake, ROS production, phagosome acidification, and intracellular bacterial killing in human monocyte-derived macrophages (MDMs) with pharmacologically inhibited cystic fibrosis transmembrane conductance regulator channel (CFTR), and improve uptake and intracellular killing of MDR P. aeruginosa in CF macrophages with impaired bactericidal activity. Moreover, ABL/PI5P stimulation of CFTR-inhibited MDM infected with MDR P. aeruginosa significantly reduces NF-κB activation and the production of TNF-α, IL-1ß, and IL-6, while increasing IL-10 and TGF-ß levels. The therapeutic efficacy of ABL/PI5P given by pulmonary administration was evaluated in a murine model of chronic infection with MDR P. aeruginosa. The treatment with ABL/PI5P significantly reduces pulmonary neutrophil infiltrate and the levels of KC and MCP-2 cytokines in the lungs, without affecting pulmonary bacterial load. Altogether, these results show that the ABL/PI5P treatment may represent a promising host-directed therapeutic approach to improve the impaired phagocytosis and to limit the potentially tissue-damaging inflammatory response in CF.


Subject(s)
Cystic Fibrosis/immunology , Immunity, Innate , Macrophages, Alveolar/immunology , Phosphatidylinositol Phosphates/pharmacology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Cystic Fibrosis/pathology , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Liposomes , Macrophages, Alveolar/pathology , Phosphatidylinositol Phosphates/pharmacokinetics , Pseudomonas Infections/pathology
6.
Sci Rep ; 10(1): 13887, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32807835

ABSTRACT

Methylglyoxal (MG) is a natural metabolite derived from glycolysis, and it inhibits the growth of cells in all kinds of organisms. We recently reported that MG inhibits nuclear division in Saccharomyces cerevisiae. However, the mechanism by which MG blocks nuclear division remains unclear. Here, we show that increase in the levels of phosphatidylinositol 3,5-bisphosphate (PtdIns(3,5)P2) is crucial for the inhibitory effects of MG on nuclear division, and the deletion of PtdIns(3,5)P2-effector Atg18 alleviated the MG-mediated inhibitory effects. Previously, we reported that MG altered morphology of the vacuole to a single swelling form, where PtdIns(3,5)P2 accumulates. The changes in the vacuolar morphology were also needed by MG to exert its inhibitory effects on nuclear division. The known checkpoint machinery, including the spindle assembly checkpoint and morphological checkpoint, are not involved in the blockade of nuclear division by MG. Our results suggest that both the accumulation of Atg18 on the vacuolar membrane and alterations in vacuolar morphology are necessary for the MG-induced inhibition of nuclear division.


Subject(s)
Autophagy-Related Proteins/metabolism , Cell Membrane/metabolism , Cell Nucleus Division/drug effects , Membrane Proteins/metabolism , Pyruvaldehyde/pharmacology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vacuoles/metabolism , Alleles , Autophagy-Related Proteins/genetics , Cell Membrane/drug effects , Membrane Proteins/genetics , Microtubules/drug effects , Microtubules/metabolism , Mutation/genetics , Phosphatidylinositol Phosphates/pharmacology , Phosphorylation/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/genetics , Spindle Poles/drug effects , Spindle Poles/metabolism , Vacuoles/drug effects
7.
Elife ; 92020 03 16.
Article in English | MEDLINE | ID: mdl-32167471

ABSTRACT

Ion selectivity is a defining feature of a given ion channel and is considered immutable. Here we show that ion selectivity of the lysosomal ion channel TPC2, which is hotly debated (Calcraft et al., 2009; Guo et al., 2017; Jha et al., 2014; Ruas et al., 2015; Wang et al., 2012), depends on the activating ligand. A high-throughput screen identified two structurally distinct TPC2 agonists. One of these evoked robust Ca2+-signals and non-selective cation currents, the other weaker Ca2+-signals and Na+-selective currents. These properties were mirrored by the Ca2+-mobilizing messenger, NAADP and the phosphoinositide, PI(3,5)P2, respectively. Agonist action was differentially inhibited by mutation of a single TPC2 residue and coupled to opposing changes in lysosomal pH and exocytosis. Our findings resolve conflicting reports on the permeability and gating properties of TPC2 and they establish a new paradigm whereby a single ion channel mediates distinct, functionally-relevant ionic signatures on demand.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channels/metabolism , Macrophages/metabolism , Raloxifene Hydrochloride/pharmacology , Animals , Benzylisoquinolines/pharmacology , Calcium/metabolism , Calcium Channel Agonists/chemistry , Calcium Channels/genetics , Fluphenazine/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , HeLa Cells , Humans , Ionomycin/pharmacology , Macrophages/drug effects , Mice , NADP/analogs & derivatives , NADP/metabolism , Phosphatidylinositol Phosphates/pharmacology , Single Molecule Imaging , Sodium/metabolism
8.
Elife ; 82019 12 11.
Article in English | MEDLINE | ID: mdl-31825310

ABSTRACT

Mammalian two-pore-channels (TPC1, 2; TPCN1, TPCN2) are ubiquitously- expressed, PI(3,5)P2-activated, Na+-selective channels in the endosomes and lysosomes that regulate luminal pH homeostasis, membrane trafficking, and Ebola viral infection. Whereas the channel activity of TPC1 is strongly dependent on membrane voltage, TPC2 lacks such voltage dependence despite the presence of the presumed 'S4 voltage-sensing' domains. By performing high-throughput screening followed by lysosomal electrophysiology, here we identified a class of tricyclic anti-depressants (TCAs) as small-molecule agonists of TPC channels. TCAs activate both TPC1 and TPC2 in a voltage-dependent manner, referred to as Lysosomal Na+ channel Voltage-dependent Activators (LyNa-VAs). We also identified another compound which, like PI(3,5)P2, activates TPC2 independent of voltage, suggesting the existence of agonist-specific gating mechanisms. Our identification of small-molecule TPC agonists should facilitate the studies of the cell biological roles of TPCs and can also readily explain the reported effects of TCAs in the modulation of autophagy and lysosomal functions.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Calcium Channel Agonists/pharmacology , Calcium Channels/metabolism , Lysosomes/enzymology , Animals , Calcium Channels/genetics , Cell Line , DNA Mutational Analysis , Humans , Mutagenesis, Site-Directed , Phosphatidylinositol Phosphates/pharmacology
9.
Mol Biol Cell ; 30(2): 201-208, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30427760

ABSTRACT

Phosphoinositides (PIs) regulate a myriad of cellular functions including membrane fusion, as exemplified by the yeast vacuole, which uses various PIs at different stages of fusion. In light of this, the effect of phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) on vacuole fusion remains unknown. PI(3,5)P2 is made by the PI3P 5-kinase Fab1 and has been characterized as a regulator of vacuole fission during hyperosmotic shock, where it interacts with the TRP Ca2+ channel Yvc1. Here we demonstrate that exogenously added dioctanoyl (C8) PI(3,5)P2 abolishes homotypic vacuole fusion. This effect was not linked to Yvc1, as fusion was equally affected using yvc1Δ vacuoles. Thus, the effects of C8-PI(3,5)P2 on fusion and fission operate through distinct mechanisms. Further testing showed that C8-PI(3,5)P2 inhibited vacuole fusion after trans-SNARE pairing. Although SNARE complex formation was unaffected, we found that C8-PI(3,5)P2 blocked outer leaflet lipid mixing. Overproduction of endogenous PI(3,5)P2 by the fab1T2250A hyperactive kinase mutant also inhibited the lipid mixing stage, bolstering the model in which PI(3,5)P2 inhibits fusion when present at elevated levels. Taken together, this work identifies a novel function for PI(3,5)P2 as a regulator of vacuolar fusion. Moreover, it suggests that this lipid acts as a molecular switch between fission and fusion.


Subject(s)
Membrane Fusion , Phosphatidylinositol Phosphates/pharmacology , SNARE Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vacuoles/metabolism , Lipids/chemistry , Membrane Fusion/drug effects , Molecular Docking Simulation , Mutation/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Saccharomyces cerevisiae Proteins/genetics , Vacuoles/drug effects
10.
Exp Cell Res ; 370(2): 601-612, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30028970

ABSTRACT

Oxysterol-binding protein (OSBP)-related proteins (ORPs) are conserved lipid binding proteins found in organisms ranging from yeast to mammals. Recent findings have indicated that these proteins mainly localize to contact sites of 2 different membranous organelles. ORP6, a member of the ORP subfamily III, is one of the least studied ORPs. Using approaches in molecular cell biology, we attempted to study the characteristics of ORP6 and found that ORP6 is abundantly expressed in mouse cultured neurons. Deconvolution microscopy of cultured cerebellar granular cells revealed that ORP6 is localized to the endoplasmic reticulum (ER) and ER-plasma membrane (PM) contact sites, where it co-localized with extended synaptotagmin2 (E-Syt2), a well-known ER-PM contact site marker. E-Syt2 also co-localized with ORP3, another subfamily III member, and ORP5, a subfamily IV member. However, ORP5 does not distribute to the same ER-PM contact sites as subfamily III members. Also, the co-expression of ORP3 but not ORP5 altered the distribution of ORP6 into the processes of cerebellar neurons. Immunoprecipitation demonstrated binding between the intermediate region of ORP6 and ORP3 or ORP6 itself. Additionally, the localization of ORP6 in the PM decreased when co-expressed with the intermediate region of ORP6, in which the pleckstrin homology (PH) domain and OSBP-related ligand binding domain (ORD) are deleted. Over-expression of this intermediate region shifted the location of a phophtidylinositol-4-phosphate (PI4P) marker from the Golgi to the PM. Knockdown of ORP6 resulted in the same shift of the PI4P marker. Collectively, our data suggests that the recruitment of ORP6 to ER-PM contact sites is involved in the turnover of PI4P in cerebellar granular neurons.


Subject(s)
Biological Transport/drug effects , Endoplasmic Reticulum/drug effects , Phosphatidylinositol Phosphates/pharmacology , Receptors, Steroid/drug effects , Biological Transport/physiology , Carrier Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mitochondrial Membranes/metabolism , Neurons/drug effects , Neurons/metabolism , Oxysterols/metabolism , Receptors, Steroid/metabolism
11.
FEBS J ; 285(6): 1146-1161, 2018 03.
Article in English | MEDLINE | ID: mdl-29405580

ABSTRACT

Saccharomyces cerevisiae possesses a transient receptor potential (TRP) channel homolog TRPY1 in its vacuolar membrane, considered to be an ancestral TRP channel. So far, studies have focused on the channel properties of TRPY1, but its regulation and physiologic role remained to be elucidated. Here, we investigated TRPY1 channel function in vitro and in vivo. Patch-clamp recording of TRPY1 in yeast vacuolar membranes showed that Ca2+ on the lumen side inhibited TRPY1-mediated channel activity, whereas luminal Zn2+ increased the currents. TRPY1 was activated in the presence of a reducing agent, 2-mercaptoethanol. The cysteine at position 624 was identified as the target for this activating action. This activation was independent of the presence of cytosolic Ca2+ . The amplitude of TRPY1-mediated current was reduced by addition of phosphatidylinositol 3-phosphate on the cytosolic side but not by phosphatidylinositol (PI) or phosphatidylinositol 3,5-phosphate. Measurement of the transient Ca2+ increase in response to hyper-osmotic shock in several yeast mutants defective in different steps of the PI phosphate biogenesis pathway supported this interpretation. Addition of a microtubule inhibitor strongly decreased the transient cytosolic Ca2+ increase upon hyper-osmotic shock. Taken together, the data indicate that the vacuolar TRPY1 Ca2+ channel mediates the perception of cytosolic signals that were induced by external changes in osmolarity, and participates in the modulation of cytosolic calcium signaling through Ca2+ release from the vacuole to maintain intracellular Ca2+ homeostasis in yeast.


Subject(s)
Calcium/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , TRPC Cation Channels/metabolism , Vacuoles/metabolism , Zinc/metabolism , Calcium/pharmacology , Cysteine/metabolism , Homeostasis , Ion Channel Gating/drug effects , Mercaptoethanol/pharmacology , Osmotic Pressure , Patch-Clamp Techniques , Phosphatidylinositol Phosphates/pharmacology , Zinc/pharmacology
12.
Sci Rep ; 7: 45120, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28345623

ABSTRACT

Phagocytosis is a key mechanism of innate immunity, and promotion of phagosome maturation may represent a therapeutic target to enhance antibacterial host response. Phagosome maturation is favored by the timely and coordinated intervention of lipids and may be altered in infections. Here we used apoptotic body-like liposomes (ABL) to selectively deliver bioactive lipids to innate cells, and then tested their function in models of pathogen-inhibited and host-impaired phagosome maturation. Stimulation of macrophages with ABLs carrying phosphatidic acid (PA), phosphatidylinositol 3-phosphate (PI3P) or PI5P increased intracellular killing of BCG, by inducing phagosome acidification and ROS generation. Moreover, ABLs carrying PA or PI5P enhanced ROS-mediated intracellular killing of Pseudomonas aeruginosa, in macrophages expressing a pharmacologically-inhibited or a naturally-mutated cystic fibrosis transmembrane conductance regulator. Finally, we show that bronchoalveolar lavage cells from patients with drug-resistant pulmonary infections increased significantly their capacity to kill in vivo acquired bacterial pathogens when ex vivo stimulated with PA- or PI5P-loaded ABLs. Altogether, these results provide the proof of concept of the efficacy of bioactive lipids delivered by ABL to enhance phagosome maturation dependent antimicrobial response, as an additional host-directed strategy aimed at the control of chronic, recurrent or drug-resistant infections.


Subject(s)
Immunity, Innate , Liposomes , Phagocytosis , Phosphatidylinositol Phosphates/immunology , Adolescent , Adult , Cell Line, Tumor , Cells, Cultured , Child , Drug Resistance, Bacterial , Female , Humans , Macrophages/drug effects , Macrophages/immunology , Male , Phagosomes/drug effects , Phagosomes/immunology , Phosphatidylinositol Phosphates/administration & dosage , Phosphatidylinositol Phosphates/pharmacology , Pseudomonas aeruginosa/immunology
13.
Cell Res ; 27(2): 253-273, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28106075

ABSTRACT

Neurite initiation is critical for neuronal morphogenesis and early neural circuit development. Recent studies showed that local actin aggregation underneath the cell membrane determined the site of neurite initiation. An immediately arising question is what signaling mechanism initiated actin aggregation. Here we demonstrate that local clustering of phosphatidylinositol 3,4-bisphosphate (PI(3,4)P2), a phospholipid with relatively few known signaling functions, is necessary and sufficient for aggregating actin and promoting neuritogenesis. In contrast, the related and more extensively studied phosphatidylinositol 4,5-bisphosphate or phosphatidylinositol (3,4,5)-trisphosphate (PIP3) molecules did not have such functions. Specifically, we showed that beads coated with PI(3,4)P2 promoted actin aggregation and neurite initiation, while pharmacological interference with PI(3,4)P2 synthesis inhibited both processes. PI(3,4)P2 clustering occurred even when actin aggregation was pharmacologically blocked, demonstrating that PI(3,4)P2 functioned as the upstream signaling molecule. Two enzymes critical for PI(3,4)P2 generation, namely, SH2 domain-containing inositol 5-phosphatase and class II phosphoinositide 3-kinase α, were complementarily and non-redundantly required for actin aggregation and neuritogenesis, as well as for subsequent dendritogenesis. Finally, we demonstrate that neural Wiskott-Aldrich syndrome protein and the Arp2/3 complex functioned downstream of PI(3,4)P2 to mediate neuritogenesis and dendritogenesis. Together, our results identify PI(3,4)P2 as an important signaling molecule during early development and demonstrate its critical role in regulating actin aggregation and neuritogenesis.


Subject(s)
Actins/metabolism , Dendrites/metabolism , Neurites/metabolism , Neurogenesis/drug effects , Phosphatidylinositol Phosphates/pharmacology , Protein Aggregates , Actin-Related Protein 2-3 Complex/metabolism , Animals , Dendrites/drug effects , Humans , Models, Biological , Neurites/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , RNA Interference , Rats, Sprague-Dawley , Wiskott-Aldrich Syndrome Protein/metabolism
14.
J Leukoc Biol ; 101(5): 1155-1168, 2017 05.
Article in English | MEDLINE | ID: mdl-28096301

ABSTRACT

Production of reactive oxygen species (ROS) in the phagosome by the NADPH oxidase is critical for mammalian immune defense against microbial infections and phosphoinositides are important regulators in this process. Phosphoinositol 3-phosphate (PI(3)P) regulates ROS production at the phagosome via p40phox by an unknown mechanism. This study tested the hypothesis that PI(3)P controls ROS production by regulating the presence of p40phox and p67phox at the phagosomal membrane. Pharmacologic inhibition of PI(3)P synthesis at the phagosome decreased the ROS production both in differentiated PLB-985 cells and human neutrophils. It also releases p67phox, the key cytosolic subunit of the oxidase, and p40phox from the phagosome. The knockdown of the PI(3)P phosphatase MTM1 or Rubicon or both increases the level of PI(3)P at the phagosome. That increase enhances ROS production inside the phagosome and triggers an extended accumulation of p67phox at the phagosome. Furthermore, the overexpression of MTM1 at the phagosomal membrane induces the disappearance of PI(3)P from the phagosome and prevents sustained ROS production. In conclusion, PI(3)P, indeed, regulates ROS production by maintaining p40phox and p67phox at the phagosomal membrane.


Subject(s)
Monocytes/immunology , NADPH Oxidases/immunology , Neutrophils/immunology , Phagosomes/immunology , Phosphatidylinositol Phosphates/immunology , Phosphoproteins/immunology , Autophagy-Related Proteins , Cell Line, Tumor , Gene Expression Regulation , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/immunology , Intracellular Membranes/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Monocytes/cytology , Monocytes/drug effects , NADPH Oxidases/genetics , Neutrophils/cytology , Neutrophils/drug effects , Phagosomes/drug effects , Phagosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol Phosphates/pharmacology , Phosphoproteins/genetics , Primary Cell Culture , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Signal Transduction
15.
Zhongguo Dang Dai Er Ke Za Zhi ; 18(8): 775-80, 2016 Aug.
Article in Chinese | MEDLINE | ID: mdl-27530799

ABSTRACT

OBJECTIVE: To investigate the effect of phosphoinositide 4-phosphate (PI4P) on human glioma U87 cells and the mechanism of action of PI4P in the development of human glioma through the overexpression or silencing of PI4P in human glioma U87 cells, and to provide a new target for basic research and clinical treatment of glioma. METHODS: LV-Helper1, LV-Helper2, pWPXLd-PI4P, and pLL3.7-shPI4P were used to package pWPXLd-PI4P and pLL3.7-shPI4P lentiviruses. The U87-GFP (PI4P-overexpression control group), U87-GFP-PI4P (PI4P-overexpression experimental group), U87-Scramble (PI4P-silencing control group), and U87-shPI4P (PI4P-silencing experimental group) cell lines were established. Wound-healing assay and Transwell assay were used to evaluate cell migration and invasion, and Western blot was used to measure the expression of PI4P in each group. RESULTS: Western blot detected the expression of exogenous PI4P in the U87-GFP-PI4P cell line, and the U87-shPI4P cell line showed reduced expression of PI4P compared with the U87-Scramble cell line in the control group. The U87-GFP-PI4P cell line with PI4P overexpression had a significantly stronger ability of migration than the U87-GFP cell line in the control group (P<0.01); the U87-shPI4P cell line with PI4P silencing had a reduced ability of migration than the U87-Scramble cell line in the control group (P<0.01). The U87 cell line with PI4P overexpression had a significantly stronger invasion ability than the control group (P<0.05); after PI4P silencing, the experimental group showed a significant reduction in invasion ability compared with the control group (P<0.05). CONCLUSIONS: In human glioma U87 cells, PI4P can promote the invasion and migration of glioma cells and may become a new target in the basic research and clinical treatment of glioma.


Subject(s)
Glioma/pathology , Phosphatidylinositol Phosphates/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Humans , Neoplasm Invasiveness
16.
Circ Res ; 119(7): 827-38, 2016 Sep 16.
Article in English | MEDLINE | ID: mdl-27514935

ABSTRACT

RATIONALE: The molecular mechanism by which ATP-binding cassette transporter A1 (ABCA1) mediates cellular binding of apolipoprotein A-I (apoA1) and nascent high-density lipoprotein (HDL) assembly is not well understood. OBJECTIVE: To determine the cell surface lipid that mediates apoA1 binding to ABCA1-expressing cells and the role it plays in nascent HDL assembly. METHODS AND RESULTS: Using multiple biochemical and biophysical methods, we found that apoA1 binds specifically to phosphatidylinositol (4,5) bis-phosphate (PIP2). Flow cytometry and PIP2 reporter-binding assays demonstrated that ABCA1 led to PIP2 redistribution from the inner to the outer leaflet of the plasma membrane. Enzymatic cleavage of cell surface PIP2 or decreased cellular PIP2 by knockdown of phosphatidylinositol-5-phosphate 4-kinase impaired apoA1 binding and cholesterol efflux to apoA1. PIP2 also increased the spontaneous solubilization of phospholipid liposomes by apoA1. Using site-directed mutagenesis, we found that ABCA1's PIP2 and phosphatidylserine translocase activities are independent from each other. Furthermore, we discovered that PIP2 is effluxed from cells to apoA1, where it is associated with HDL in plasma, and that PIP2 on HDL is taken up by target cells in a scavenger receptor-BI-dependent manner. Mouse plasma PIP2 levels are apoA1 gene dosage-dependent and are >1 µM in apoA1 transgenic mice. CONCLUSIONS: ABCA1 has PIP2 floppase activity, which increases cell surface PIP2 levels that mediate apoA1 binding and lipid efflux during nascent HDL assembly. We found that PIP2 itself is effluxed to apoA1 and it circulates on plasma HDL, where it can be taken up via the HDL receptor scavenger receptor-BI.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Apolipoprotein A-I/metabolism , Cell Membrane/metabolism , Lipoproteins, HDL/metabolism , Phosphatidylinositol Phosphates/metabolism , Animals , Biological Transport/physiology , Cricetinae , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphatidylinositol Phosphates/pharmacology , Protein Binding/physiology
17.
J Drug Target ; 23(7-8): 698-709, 2015.
Article in English | MEDLINE | ID: mdl-26453165

ABSTRACT

BACKGROUND: Phosphatidylinositol-3,4,5-trisphosphate (PIP3) is a major lipid second messenger in insulin-mediated signalling towards the metabolic actions of this hormone in muscle and fat. PURPOSE: Assessing the intracellular transport of exogenous PIP3 attached to a polymeric carrier in an attempt to overcome cellular insulin resistance. METHODS: Artificial chromatic bio-mimetic membrane vesicles composed of dimyristoylphosphatidylcholine and polydiacetylene were applied to screen the polymeric carriers. PIP3 cellular localization and bio-activity was assessed by fluorescent and live-cell microscopy in L6 muscle cells and in 3T3-L1 adipocytes. RESULTS AND DISCUSSION: We demonstrate that a specific-branched polyethylenimine (PEI-25, 25 kDa) carrier forms complexes with PIP3 that interact with the bio-mimetic membrane vesicles in a manner predictive of their interaction with cells: In L6 muscle cells, PEI-25/fluorescent-PIP3 complexes are retarded at the cell perimeter. PEI-25/PIP3 complexes retain their bio-activity, engaging signalling steps downstream of PIP3, even in muscle cells rendered insulin resistant by exposure to high glucose/high insulin. CONCLUSIONS: Inducing insulin actions by intracellular PIP3 delivery (PEI-25/PIP3 complexes) in some forms of insulin-resistant cells provides the first proof-of-principle for the potential therapeutic use of PIP3 in a "second-messenger agonist" approach. In addition, utilization of an artificial bio-mimetic membrane platform to screen for highly efficient PIP3 delivery predicts biological function in cells.


Subject(s)
Drug Delivery Systems , Insulin Resistance , Insulin/metabolism , Phosphatidylinositol Phosphates/administration & dosage , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cells, Cultured , Drug Carriers/chemistry , Mice , Myoblasts/drug effects , Myoblasts/metabolism , Phosphatidylinositol Phosphates/pharmacology , Polyethyleneimine/chemistry , Polymers/chemistry , Rats , Signal Transduction/drug effects
18.
Cell Signal ; 27(7): 1488-98, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25797048

ABSTRACT

Phosphatidylinositol trisphosphate (PIP3) has been implicated in many platelet functions however many of the mechanisms need clarification. We have used cell permeable analogues of PIP3,1-O-(1,2-di-palmitoyl-sn-glyero-3-O-phosphoryl)-D-myo-inositol-3,4,5-trisphosphate (DiC16-PIP3) or 1-O-(1,2-di-octanoyl-sn-glyero-3-O-phosphoryl)-D-myo-inositol-3,4,5-trisphosphate (DiC8-PIP3) to study their effects on activation on washed human platelets. Addition of either DiC8- or DiC16-PIP3 to human platelets induced aggregation in the presence of extracellular Ca(2+). This was reduced by the presence of indomethacin, the phospholipase C inhibitor U73122 and apyrase. DiC8-PIP3 induced the phosphorylation of Akt-Ser(473) which was reduced by the Akt inhibitor IV, wortmannin and EGTA (suggesting a dependence on Ca(2+) entry). In Fura2 loaded platelets DiC8-PIP3 was effective at increasing intracellular Ca(2+) in a distinct and transient manner that was reduced in the presence of indomethacin, U73122 and 2-aminoethyl diphenylborinate (2APB). Ca(2+) elevation was reduced by the non-SOCE inhibitor LOE908 and also by the SOCE inhibitor BTP2. DiC8-PIP3 induced the release of Ca(2+) from stores which was not affected by the proton dissipating agent bafilomycin A1 and was more potent than the two-pore channel agonist DiC8-PI[3,5]P2 suggesting release from an endoplasmic reticulum type store. DiC8-PIP3 weakly induced the tyrosine phosphorylation of Syk but not of PLCγ2. Finally like thrombin DiC8-PIP3 induced the formation of thromboxane B2 that was inhibited by the Akt inhibitor IV. These studies suggest that PIP3 via Ca(2+) elevation and Akt phosphorylation forms a central role in thromboxane A2 formation and the amplification of platelet activation.


Subject(s)
Blood Platelets/drug effects , Calcium/metabolism , Phosphatidylinositol Phosphates/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Thromboxane A2/metabolism , Androstadienes/pharmacology , Blood Platelets/cytology , Blood Platelets/metabolism , Egtazic Acid/pharmacology , Enzyme-Linked Immunosorbent Assay , Fura-2/chemistry , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Syk Kinase , Thromboxane A2/analysis , Wortmannin
19.
Cell Calcium ; 56(6): 446-56, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25266962

ABSTRACT

Mucolipin synthetic agonist 1 (ML-SA1) was recently identified to activate mammalian TRPML channels and shown to alleviate lipid accumulation in lysosomes of cellular models of lysosome storage diseases, mucolipidosis type IV (MLIV) and Niemann-Pick's disease type C (NPC). Owning to its potential use in complimenting genetic studies in Drosophila melanogaster to elucidate the cellular and physiological functions of TRPML channels, we examined the effect of ML-SA1 on Drosophila TRPML expressed in HEK293 cells using whole-cell, inside-out, and whole-lysosome electrophysiological recordings. We previously showed that when expressed in HEK293 cells, Drosophila TRPML was localized and functional on both plasma membrane and endolysosome. We show here that in both inside-out patches excised from the plasma membrane and whole-lysosome recordings from enlarged endolysosome vacuoles, ML-SA1 failed to activate TRPML unless exogenous phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2] was applied. At 1 µM ML-SA1, the sensitivity of TRPML to PI(3,5)P2 increased approximately by 10-fold and at 10 µM ML-SA1, the deactivation of PI(3,5)P2-evoked TRPML currents was markedly slowed. On the other hand, constitutive activation of TRPML by a mutation that mimics the varitint-waddler (Va) mutation of mouse TRPML3 rendered the insect channel sensitive to activation by ML-SA1 alone. Moreover, different from the insect TRPML, mouse TRPML1 was readily activated by ML-SA1 independent of PI(3,5)P2. Thus, our data reveal that while ML-SA1 acts as a true agonist at mouse TRPML1, it behaves as an allosteric activator of the Drosophila TRPML, showing dependence on and the ability to stabilize open conformation of the insect channels.


Subject(s)
Drosophila Proteins/drug effects , Transient Receptor Potential Channels/drug effects , Animals , Cell Membrane/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , HEK293 Cells , Humans , Lipid Metabolism/drug effects , Mice , Patch-Clamp Techniques , Phosphatidylinositol Phosphates/pharmacology , Phthalimides/pharmacology , Quinolines/pharmacology , Species Specificity , Transient Receptor Potential Channels/metabolism
20.
Nat Chem Biol ; 10(9): 753-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108821

ABSTRACT

Phosphatidylinositol bisphosphate (PIP2) regulates the activities of many membrane proteins, including ion channels, through direct interactions. However, the affinity of PIP2 is so high for some channel proteins that its physiological role as a modulator has been questioned. Here we show that PIP2 is a key cofactor for activation of small conductance Ca2+-activated potassium channels (SKs) by Ca(2+)-bound calmodulin (CaM). Removal of the endogenous PIP2 inhibits SKs. The PIP2-binding site resides at the interface of CaM and the SK C terminus. We further demonstrate that the affinity of PIP2 for its target proteins can be regulated by cellular signaling. Phosphorylation of CaM T79, located adjacent to the PIP2-binding site, by casein kinase 2 reduces the affinity of PIP2 for the CaM-SK channel complex by altering the dynamic interactions among amino acid residues surrounding the PIP2-binding site. This effect of CaM phosphorylation promotes greater channel inhibition by G protein-mediated hydrolysis of PIP2.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Phosphatidylinositol Phosphates/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Acetylcholine/pharmacology , Amino Acids/metabolism , Binding Sites , Casein Kinase II/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/pharmacology , HEK293 Cells , Humans , Models, Molecular , Phosphatidylinositol Phosphates/physiology , Phosphorylation , Protein Conformation , Protein Kinase C/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...