Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 42(10): e112234, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36970857

ABSTRACT

The interferon-induced transmembrane proteins (IFITM) are implicated in several biological processes, including antiviral defense, but their modes of action remain debated. Here, taking advantage of pseudotyped viral entry assays and replicating viruses, we uncover the requirement of host co-factors for endosomal antiviral inhibition through high-throughput proteomics and lipidomics in cellular models of IFITM restriction. Unlike plasma membrane (PM)-localized IFITM restriction that targets infectious SARS-CoV2 and other PM-fusing viral envelopes, inhibition of endosomal viral entry depends on lysines within the conserved IFITM intracellular loop. These residues recruit Phosphatidylinositol 3,4,5-trisphosphate (PIP3) that we show here to be required for endosomal IFITM activity. We identify PIP3 as an interferon-inducible phospholipid that acts as a rheostat for endosomal antiviral immunity. PIP3 levels correlated with the potency of endosomal IFITM restriction and exogenous PIP3 enhanced inhibition of endocytic viruses, including the recent SARS-CoV2 Omicron variant. Together, our results identify PIP3 as a critical regulator of endosomal IFITM restriction linking it to the Pi3K/Akt/mTORC pathway and elucidate cell-compartment-specific antiviral mechanisms with potential relevance for the development of broadly acting antiviral strategies.


Subject(s)
Antiviral Agents , COVID-19 , Humans , Interferons/metabolism , Phospholipids , Phosphatidylinositol 3-Kinases/metabolism , RNA, Viral , RNA-Binding Proteins/metabolism , SARS-CoV-2/metabolism , Virus Internalization , Membrane Proteins/metabolism
2.
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
3.
Proc Natl Acad Sci U S A ; 120(19): e2218906120, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37126708

ABSTRACT

Cellular sensing of most environmental cues involves receptors that affect a signal-transduction excitable network (STEN), which is coupled to a cytoskeletal excitable network (CEN). We show that the mechanism of sensing of nanoridges is fundamentally different. CEN activity occurs preferentially on nanoridges, whereas STEN activity is constrained between nanoridges. In the absence of STEN, waves disappear, but long-lasting F-actin puncta persist along the ridges. When CEN is suppressed, wave propagation is no longer constrained by nanoridges. A computational model reproduces these experimental observations. Our findings indicate that nanotopography is sensed directly by CEN, whereas STEN is only indirectly affected due to a CEN-STEN feedback loop. These results explain why texture sensing is robust and acts cooperatively with multiple other guidance cues in complex, in vivo microenvironments.


Subject(s)
Actin Cytoskeleton , Cytoskeleton , Cell Movement , Actins , Microtubules
4.
Biochem Soc Trans ; 52(4): 1785-1794, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38934501

ABSTRACT

Macropinocytosis is a broadly conserved endocytic process discovered nearly 100 years ago, yet still poorly understood. It is prominent in cancer cell feeding, immune surveillance, uptake of RNA vaccines and as an invasion route for pathogens. Macropinocytic cells extend large cups or flaps from their plasma membrane to engulf droplets of medium and trap them in micron-sized vesicles. Here they are digested and the products absorbed. A major problem - discussed here - is to understand how cups are shaped and closed. Recently, lattice light-sheet microscopy has given a detailed description of this process in Dictyostelium amoebae, leading to the 'stalled-wave' model for cup formation and closure. This is based on membrane domains of PIP3 and active Ras and Rac that occupy the inner face of macropinocytic cups and are readily visible with suitable reporters. These domains attract activators of dendritic actin polymerization to their periphery, creating a ring of protrusive F-actin around themselves, thus shaping the walls of the cup. As domains grow, they drive a wave of actin polymerization across the plasma membrane that expands the cup. When domains stall, continued actin polymerization under the membrane, combined with increasing membrane tension in the cup, drives closure at lip or base. Modelling supports the feasibility of this scheme. No specialist coat proteins or contractile activities are required to shape and close cups: rings of actin polymerization formed around PIP3 domains that expand and stall seem sufficient. This scheme may be widely applicable and begs many biochemical questions.


Subject(s)
Cell Membrane , Dictyostelium , Pinocytosis , Pinocytosis/physiology , Cell Membrane/metabolism , Humans , Actins/metabolism , Animals , Models, Biological
5.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Article in English | MEDLINE | ID: mdl-34385319

ABSTRACT

The protein kinase Akt is one of the primary effectors of growth factor signaling in the cell. Akt responds specifically to the lipid second messengers phosphatidylinositol-3,4,5-trisphosphate [PI(3,4,5)P3] and phosphatidylinositol-3,4-bisphosphate [PI(3,4)P2] via its PH domain, leading to phosphorylation of its activation loop and the hydrophobic motif of its kinase domain, which are critical for activity. We have now determined the crystal structure of Akt1, revealing an autoinhibitory interface between the PH and kinase domains that is often mutated in cancer and overgrowth disorders. This interface persists even after stoichiometric phosphorylation, thereby restricting maximum Akt activity to PI(3,4,5)P3- or PI(3,4)P2-containing membranes. Our work helps to resolve the roles of lipids and phosphorylation in the activation of Akt and has wide implications for the spatiotemporal control of Akt and potentially lipid-activated kinase signaling in general.


Subject(s)
Phosphatidylinositol Phosphates/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Animals , Binding Sites , Humans , Insecta , Lipid Metabolism , Phosphatidylinositol Phosphates/genetics , Protein Binding , Protein Conformation , Protein Domains , Proto-Oncogene Proteins c-akt/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Sf9 Cells
6.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443153

ABSTRACT

The differentiation of cells depends on a precise control of their internal organization, which is the result of a complex dynamic interplay between the cytoskeleton, molecular motors, signaling molecules, and membranes. For example, in the developing neuron, the protein ADAP1 (ADP-ribosylation factor GTPase-activating protein [ArfGAP] with dual pleckstrin homology [PH] domains 1) has been suggested to control dendrite branching by regulating the small GTPase ARF6. Together with the motor protein KIF13B, ADAP1 is also thought to mediate delivery of the second messenger phosphatidylinositol (3,4,5)-trisphosphate (PIP3) to the axon tip, thus contributing to PIP3 polarity. However, what defines the function of ADAP1 and how its different roles are coordinated are still not clear. Here, we studied ADAP1's functions using in vitro reconstitutions. We found that KIF13B transports ADAP1 along microtubules, but that PIP3 as well as PI(3,4)P2 act as stop signals for this transport instead of being transported. We also demonstrate that these phosphoinositides activate ADAP1's enzymatic activity to catalyze GTP hydrolysis by ARF6. Together, our results support a model for the cellular function of ADAP1, where KIF13B transports ADAP1 until it encounters high PIP3/PI(3,4)P2 concentrations in the plasma membrane. Here, ADAP1 disassociates from the motor to inactivate ARF6, promoting dendrite branching.


Subject(s)
ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Nerve Tissue Proteins/metabolism , Phosphatidylinositols/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Axons/metabolism , Biological Transport/physiology , Cell Membrane/metabolism , Cytoskeleton/metabolism , GTPase-Activating Proteins/metabolism , Humans , Inositol Phosphates/metabolism , Kinesins/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/physiology , Phosphatidylinositol Phosphates/metabolism , Signal Transduction
7.
Int J Mol Sci ; 25(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38339058

ABSTRACT

Given the role of phosphatidylinositol 3,4,5-trisphosphate (PIP3) in modulating cellular processes such as proliferation, survival, and migration, we hypothesized its potential as a novel therapeutic agent for wound closure enhancement. In this study, PIP3 was examined in its free form or as a complex with cationic starch (Q-starch) as a carrier. The intracellular bioactivity and localization of free PIP3 and the Q-starch/PIP3 complexes were examined. Our results present the capability of Q-starch to form complexes with PIP3, facilitate its cellular membrane internalization, and activate intracellular paths leading to enhanced wound healing. Both free PIP3 and Q-starch/PIP3 complexes enhanced monolayer gap closure in scratch assays and induced amplified collagen production within HaCAT and BJ fibroblast cells. Western blot presented enhanced AKT activation by free or complexed PIP3 in BJ fibroblasts in which endogenous PIP3 production was pharmacologically inhibited. Furthermore, both free PIP3 and Q-starch/PIP3 complexes expedited wound closure in mice, after single or daily dermal injections into the wound margins. Free PIP3 and the Q-starch/PIP3 complexes inherently activated the AKT signaling pathway, which is responsible for crucial wound healing processes such as migration; this was also observed in wound assays in mice. PIP3 was identified as a promising molecule for enhancing wound healing, and its ability to circumvent PI3K inhibition suggests possible implications for chronic wound healing.


Subject(s)
Proto-Oncogene Proteins c-akt , Wound Healing , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Wound Healing/physiology , Signal Transduction/physiology , Fibroblasts/metabolism , Starch/metabolism , Cell Proliferation/physiology
8.
Subcell Biochem ; 98: 41-59, 2022.
Article in English | MEDLINE | ID: mdl-35378702

ABSTRACT

Macropinocytosis is a relatively unexplored form of large-scale endocytosis driven by the actin cytoskeleton. Dictyostelium amoebae form macropinosomes from cups extended from the plasma membrane, then digest their contents and absorb the nutrients in the endo-lysosomal system. They use macropinocytosis for feeding, maintaining a high rate of fluid uptake that makes assay and experimentation easy. Mutants collected over the years identify cytoskeletal and signalling proteins required for macropinocytosis. Cups are organized around plasma membrane domains of intense PIP3, Ras and Rac signalling, proper formation of which also depends on the RasGAPs NF1 and RGBARG, PTEN, the PIP3-regulated protein kinases Akt and SGK and their activators PDK1 and TORC2, Rho proteins, plus other components yet to be identified. This PIP3 domain directs dendritic actin polymerization to the extending lip of macropinocytic cups by recruiting a ring of the SCAR/WAVE complex around itself and thus activating the Arp2/3 complex. The dynamics of PIP3 domains are proposed to shape macropinocytic cups from start to finish. The role of the Ras-PI3-kinase module in organizing feeding structures in unicellular organisms most likely predates its adoption into growth factor signalling, suggesting an evolutionary origin for growth factor signalling.


Subject(s)
Amoeba , Dictyostelium , Actin Cytoskeleton/metabolism , Amoeba/metabolism , Dictyostelium/genetics , Dictyostelium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pinocytosis
9.
Semin Cell Dev Biol ; 100: 133-142, 2020 04.
Article in English | MEDLINE | ID: mdl-31836289

ABSTRACT

The directed movements of individual, groups, or sheets of cells at specific times in particular locations bring about form and complexity to developing organisms. Cells move by extending protrusions, such as macropinosomes, pseudopods, lamellipods, filopods, or blebs. Although many of the cytoskeletal components within these structures are known, less is known about the mechanisms that determine their location, number, and characteristics. Recent evidence suggests that control may be exerted by a signal transduction excitable network whose components and activities, including Ras, PI3K, TorC2, and phosphoinositides, self-organize on the plasma membrane and propagate in waves. The waves drive the various types of protrusions, which in turn, determine the modes of cell migration. Acute perturbations at specific points in the network produce abrupt shifts in protrusion type, including transitions from pseudopods to filopods or lamellipods. These observations have also contributed to a delineation of the signal transduction network, including candidate fast positive and delayed negative feedback loops. The network contains many oncogenes and tumor suppressors, and other molecules which have recently been implicated in developmental and metabolic abnormalities. Thus, the concept of signal transduction network excitability in cell migration can be used to understand disease states and morphological changes occurring in development.


Subject(s)
Cell Movement , Disease , Metabolic Networks and Pathways , Actins/metabolism , Animals , Cytoskeleton/metabolism , Humans , Signal Transduction
10.
FASEB J ; 35(4): e21508, 2021 04.
Article in English | MEDLINE | ID: mdl-33710706

ABSTRACT

Migrating tumor cells are characterized by a sustained front-rear asymmetry, with a front enriched in filamentous actin, which is induced by Rho small GTPase Rac. Regulation of Rac activity by its regulators should be required for effective motility. Here, we show that FilGAP, a GTPase-activating protein (GAP) for Rac, controls front-rear polarity and contributes to maintain effective tumor cell migration through the extracellular matrix (ECM). Overexpression of FilGAP in breast cancer cells induced polarized morphology and led to increased migration speed in collagen matrices, while depletion of FilGAP impaired the cell polarity and migration. FilGAP localizes to the cell front through its pleckstrin-homology (PH) domain in a phosphatidylinositol 3,4,5-trisphosphate (PIP3)-dependent manner and appears to inactivate Rac at its site. We found that the affinity of PH domain to PIP3 is critically involved in the maintenance of cell polarity. Moreover, small GTPase ADP-ribosylation factor 6 (Arf6), which binds to the FilGAP PH domain, also regulates FilGAP-mediated cell polarity and migration of breast cancer cells. We propose that FilGAP regulates front-rear polarity through its PIP3 and Arf6 binding in tumor cell migration through the ECM.


Subject(s)
Cell Movement/physiology , Cell Polarity/physiology , GTPase-Activating Proteins/metabolism , ADP-Ribosylation Factor 6 , Actin Cytoskeleton/metabolism , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , rho-Associated Kinases/metabolism
11.
Proc Natl Acad Sci U S A ; 116(22): 10798-10803, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31076553

ABSTRACT

The transformation of molecular binding events into cellular decisions is the basis of most biological signal transduction. A fundamental challenge faced by these systems is that reliance on protein-ligand chemical affinities alone generally results in poor sensitivity to ligand concentration, endangering the system to error. Here, we examine the lipid-binding pleckstrin homology and Tec homology (PH-TH) module of Bruton's tyrosine kinase (Btk). Using fluorescence correlation spectroscopy (FCS) and membrane-binding kinetic measurements, we identify a phosphatidylinositol (3-5)-trisphosphate (PIP3) sensing mechanism that achieves switch-like sensitivity to PIP3 levels, surpassing the intrinsic affinity discrimination of PIP3:PH binding. This mechanism employs multiple PIP3 binding as well as dimerization of Btk on the membrane surface. Studies in live cells confirm that mutations at the dimer interface and peripheral site produce effects comparable to that of the kinase-dead Btk in vivo. These results demonstrate how a single protein module can institute an allosteric counting mechanism to achieve high-precision discrimination of ligand concentration. Furthermore, this activation mechanism distinguishes Btk from other Tec family member kinases, Tec and Itk, which we show are not capable of dimerization through their PH-TH modules. This suggests that Btk plays a critical role in the stringency of the B cell response, whereas T cells rely on other mechanisms to achieve stringency.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/chemistry , Agammaglobulinaemia Tyrosine Kinase/metabolism , Signal Transduction/physiology , Animals , B-Lymphocytes/metabolism , Cell Line , Chickens , Mice , Models, Molecular , Mutation , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Protein Conformation , Protein Domains/physiology , Protein Multimerization
12.
Proc Natl Acad Sci U S A ; 116(19): 9390-9399, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31019091

ABSTRACT

Bruton's tyrosine kinase (Btk) is critical for B cell proliferation and activation, and the development of Btk inhibitors is a vigorously pursued strategy for the treatment of various B cell malignancies. A detailed mechanistic understanding of Btk activation has, however, been lacking. Here, inspired by a previous suggestion that Btk activation might depend on dimerization of its lipid-binding PH-TH module on the cell membrane, we performed long-timescale molecular dynamics simulations of membrane-bound PH-TH modules and observed that they dimerized into a single predominant conformation. We found that the phospholipid PIP3 stabilized the dimer allosterically by binding at multiple sites, and that the effects of PH-TH mutations on dimer stability were consistent with their known effects on Btk activity. Taken together, our simulation results strongly suggest that PIP3-mediated dimerization of Btk at the cell membrane is a critical step in Btk activation.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/chemistry , Agammaglobulinaemia Tyrosine Kinase/metabolism , Cell Membrane/enzymology , Agammaglobulinaemia Tyrosine Kinase/genetics , Binding Sites , Cell Membrane/chemistry , Cell Membrane/genetics , Dimerization , Enzyme Activation , Humans , Molecular Dynamics Simulation , Mutation , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Phosphorylation
13.
Int J Mol Sci ; 23(17)2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36077184

ABSTRACT

Phospholipid scramblase 4 (PLSCR4) is a member of a conserved enzyme family with high relevance for the remodeling of phospholipid distribution in the plasma membrane and the regulation of cellular signaling. While PLSCR1 and -3 are involved in the regulation of adipose-tissue expansion, the role of PLSCR4 is so far unknown. PLSCR4 is significantly downregulated in an adipose-progenitor-cell model of deficiency for phosphatase and tensin homolog (PTEN). PTEN acts as a tumor suppressor and antagonist of the growth and survival signaling phosphoinositide 3-kinase (PI3K)/AKT cascade by dephosphorylating phosphatidylinositol-3,4,5-trisphosphate (PIP3). Patients with PTEN germline deletion frequently develop lipomas. The underlying mechanism for this aberrant adipose-tissue growth is incompletely understood. PLSCR4 is most highly expressed in human adipose tissue, compared with other phospholipid scramblases, suggesting a specific role of PLSCR4 in adipose-tissue biology. In cell and mouse models of lipid accumulation, we found PLSCR4 to be downregulated. We observed increased adipogenesis in PLSCR4-knockdown adipose progenitor cells, while PLSCR4 overexpression attenuated lipid accumulation. PLSCR4 knockdown was associated with increased PIP3 levels and the activation of AKT. Our results indicated that PLSCR4 is a regulator of PI3K/AKT signaling and adipogenesis and may play a role in PTEN-associated adipose-tissue overgrowth and lipoma formation.


Subject(s)
Phosphatidylinositol 3-Kinases , Phospholipid Transfer Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adipocytes/metabolism , Animals , Humans , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols , Phospholipid Transfer Proteins/genetics
14.
J Lipid Res ; 62: 100081, 2021.
Article in English | MEDLINE | ID: mdl-33933440

ABSTRACT

Nuclear receptors are transcription factors that bind lipids, an event that induces a structural conformation of the receptor that favors interaction with transcriptional coactivators. The nuclear receptor steroidogenic factor-1 (SF-1, NR5A1) binds the signaling phosphoinositides PI(4,5)P2 (PIP2) and PI(3,4,5)P3 (PIP3), and our previous crystal structures showed how the phosphoinositide headgroups regulate SF-1 function. However, what role the acyl chains play in regulating SF-1 structure remains unaddressed. Here, we used X-ray crystallography with in vitro binding and functional assays to examine how the acyl chains of PIP3 regulate human SF-1 ligand-binding domain structure and function. Altering acyl chain length and unsaturation regulates apparent binding of all tested phosphoinositides to SF-1. Mass spectrometry-based lipidomics data suggest C16 and C18 phospholipids preferentially associate with SF-1 expressed ectopically in bacteria. We then solved the 2.5 Å crystal structure of SF-1 bound to dioleoyl PIP3(18:1/18:1) to compare it with a matched structure of SF-1 bound to dipalmitoyl PIP3(16:0/16:0). The dioleoyl-bound structure was severely disordered in a specific SF-1 region associated with pathogenic human polymorphisms and within the coactivator-binding region critical for SF-1 function while inducing increased sensitivity to protease digestion in solution. Validating these structural observations, in vitro functional studies showed dioleoyl PIP3 induced 6-fold poorer affinity of a peroxisome proliferator-activated receptor gamma coactivator 1-alpha coactivator peptide for SF-1 compared with dipalmitoyl PIP3. Together, these data suggest the chemical nature of the phosphoinositide acyl chains controls the ordered state of specific, clinically important structural regions in SF-1, regulating SF-1 function in vitro.


Subject(s)
Phosphatidylinositols
15.
J Cell Sci ; 132(20)2019 10 22.
Article in English | MEDLINE | ID: mdl-31540955

ABSTRACT

Akt signalling is central to cell survival, metabolism, protein and lipid homeostasis, and is impaired in Parkinson's disease (PD). Akt activation is reduced in the brain in PD, and by many PD-causing genes, including PINK1 This study investigated the mechanisms by which PINK1 regulates Akt signalling. Our results reveal for the first time that PINK1 constitutively activates Akt in a PINK1-kinase dependent manner in the absence of growth factors, and enhances Akt activation in normal growth medium. In PINK1-modified MEFs, agonist-induced Akt signalling failed in the absence of PINK1, due to PINK1 kinase-dependent increases in PI(3,4,5)P3 at both plasma membrane and Golgi being significantly impaired. In the absence of PINK1, PI(3,4,5)P3 levels did not increase in the Golgi, and there was significant Golgi fragmentation, a recognised characteristic of PD neuropathology. PINK1 kinase activity protected the Golgi from fragmentation in an Akt-dependent fashion. This study demonstrates a new role for PINK1 as a primary upstream activator of Akt via PINK1 kinase-dependent regulation of its primary activator PI(3,4,5)P3, providing novel mechanistic information on how loss of PINK1 impairs Akt signalling in PD.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cell Membrane/metabolism , Golgi Apparatus/metabolism , Parkinson Disease/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Membrane/genetics , Golgi Apparatus/genetics , Humans , Mice , Mice, Knockout , Parkinson Disease/genetics , Phosphatidylinositol Phosphates/genetics , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
16.
Pharmacol Res ; 165: 105415, 2021 03.
Article in English | MEDLINE | ID: mdl-33412279

ABSTRACT

Genetically modified animal studies have revealed specific expression patterns and unequivocal roles of class I PI3K isoenzymes. PI3K(p110α), a catalytic subunit of class I PI3Ks is ubiquitously expressed and is well characterised in the cardiovascular system. Given that genetic inhibition of PI3K(p110α) causes lethal phenotype embryonically, the catalytic subunit is critically important in housekeeping and biological processes. A growing number of studies underpin crucial roles of PI3K(p110α) in cell survival, proliferation, hypertrophy and arrhythmogenesis. While the studies provide great insights, the precise mechanisms involved in PI3K(p110α) hypofunction and atrial fibrillation (AF) are not fully known. AF is a well recognised clinical problem with significant management limitations. In this translational review, we attempted a narration of PI3K(p110α) hypofunction in the molecular basis of AF pathophysiology. We sought to cautiously highlight the relevance of this molecule in the therapeutic approaches for AF management per se (i.e without conditions associate with cell proliferation, like cancer), and in mitigating effects of clinical risk factors in atrial substrate formation leading to AF progression. We also considered PI3K(p110α) in AF gene association, with the aim of identifying mechanistic links between the ever increasingly well-defined genetic loci (regions and genes) and AF. Such mechanisms will aid in identifying new drug targets for arrhythmogenic substrate and AF.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/physiopathology , Class Ia Phosphatidylinositol 3-Kinase/physiology , Animals , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/etiology , Class Ia Phosphatidylinositol 3-Kinase/drug effects , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Humans
17.
Semin Cancer Biol ; 59: 50-65, 2019 12.
Article in English | MEDLINE | ID: mdl-30922959

ABSTRACT

Phosphoinositides are a group of lipids that regulate intracellular signaling and subcellular biological events. The signaling by phosphatidylinositol-3,4,5-trisphosphate and Akt mediates the action of growth factors that are essential for cell proliferation, gene transcription, cell migration, and polarity. The hyperactivation of this signaling has been identified in different cancer cells; and, it has been implicated in oncogenic transformation and cancer cell malignancy. Recent studies have argued the role of phosphoinositides in cancer cell dynamics, including actin cytoskeletal rearrangement at the plasma membrane and the organization of intracellular compartments. The focus of this review is to summarize the impact of the activities of phosphoinositide phosphatases on intracellular signaling related to cancer cell dynamics and to discuss how the abnormalities in the activities of the enzymes alter the levels of phosphoinositides in cancer cells.


Subject(s)
Neoplasms/etiology , Neoplasms/metabolism , Phosphoinositide Phosphatases/metabolism , Animals , Biomarkers , Energy Metabolism , Humans , Neoplasms/drug therapy , Neoplasms/pathology , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide Phosphatases/genetics , Signal Transduction
18.
J Biol Chem ; 293(24): 9292-9300, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29735527

ABSTRACT

Defects in phosphatase and tensin homolog (PTEN) are associated with neurological disorders and tumors. PTEN functions at two primary intracellular locations: the plasma membrane and the nucleus. At the membrane, PTEN functions as a phosphatidylinositol (3,4,5)-trisphosphate phosphatase and suppresses PI 3-kinase signaling that drives cell growth and tumorigenesis. However, the in vivo function of nuclear PTEN is unclear. Here, using CRISPR/Cas9, we generated a mouse model in which PTEN levels in the nucleus are decreased. Nuclear PTEN-deficient mice were born with microcephaly and maintained a small brain during adulthood. The size of neuronal soma was significantly smaller in the cerebellum, cerebral cortex, and hippocampus. Also, these mice were prone to seizure. No changes in PI 3-kinase signaling were observed. By contrast, the size of other organs was unaffected. Therefore, nuclear PTEN is essential for the health of the brain by promoting the growth of neuronal soma size during development.


Subject(s)
CRISPR-Cas Systems , Cell Nucleus/genetics , Microcephaly/genetics , Neurons/pathology , PTEN Phosphohydrolase/genetics , Seizures/genetics , Amino Acid Substitution , Animals , Brain/metabolism , Brain/pathology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Size , Female , Gene Editing , Male , Mice , Microcephaly/complications , Microcephaly/pathology , Mutation , Neurons/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Seizures/complications , Seizures/pathology , Signal Transduction
19.
Biochem J ; 475(3): 643-648, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29444849

ABSTRACT

In a role distinct from and perhaps more ancient than that in signal transduction, PIP3 and Ras help to spatially organize the actin cytoskeleton into macropinocytic cups. These large endocytic structures are extended by actin polymerization from the cell surface and have at their core an intense patch of active Ras and PIP3, around which actin polymerizes, creating cup-shaped projections. We hypothesize that active Ras and PIP3 self-amplify within macropinocytic cups, in a way that depends on the structural integrity of the cup. Signalling that triggers macropinocytosis may therefore be amplified downstream in a way that depends on macropinocytosis. This argument provides a context for recent findings that signalling to Akt (an effector of PIP3) is sensitive to cytoskeletal and macropinocytic inhibitors.


Subject(s)
Actin Cytoskeleton/genetics , Pinocytosis/genetics , Proto-Oncogene Proteins c-akt/genetics , Actin Cytoskeleton/chemistry , Cell Membrane/genetics , Dictyostelium/genetics , Humans , Proto-Oncogene Proteins c-akt/chemistry , Signal Transduction/genetics
20.
Proc Natl Acad Sci U S A ; 113(46): 13069-13074, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27799542

ABSTRACT

Protein kinase A (PKA) phosphorylates Gli proteins, acting as a negative regulator of the Hedgehog pathway. PKA was recently detected within the cilium, and PKA activity specifically in cilia regulates Gli processing. Using a cilia-targeted genetically encoded sensor, we found significant basal PKA activity. Using another targeted sensor, we measured basal ciliary cAMP that is fivefold higher than whole-cell cAMP. The elevated basal ciliary cAMP level is a result of adenylyl cyclase 5 and 6 activity that depends on ciliary phosphatidylinositol (3,4,5)-trisphosphate (PIP3), not stimulatory G protein (Gαs), signaling. Sonic Hedgehog (SHH) reduces ciliary cAMP levels, inhibits ciliary PKA activity, and increases Gli1. Remarkably, SHH regulation of ciliary cAMP and downstream signals is not dependent on inhibitory G protein (Gαi/o) signaling but rather Ca2+ entry through a Gd3+-sensitive channel. Therefore, PIP3 sustains high basal cAMP that maintains PKA activity in cilia and Gli repression. SHH activates Gli by inhibiting cAMP through a G protein-independent mechanism that requires extracellular Ca2+ entry.


Subject(s)
Calcium/metabolism , Cilia/metabolism , Cyclic AMP/metabolism , Hedgehog Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Animals , Cell Line , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblasts/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL