Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.600
Filter
1.
Elife ; 122024 Jun 04.
Article in English | MEDLINE | ID: mdl-38831696

ABSTRACT

During macroautophagy, cytoplasmic constituents are engulfed by autophagosomes. Lysosomes fuse with closed autophagosomes but not with unclosed intermediate structures. This is achieved in part by the late recruitment of the autophagosomal SNARE syntaxin 17 (STX17) to mature autophagosomes. However, how STX17 recognizes autophagosome maturation is not known. Here, we show that this temporally regulated recruitment of STX17 depends on the positively charged C-terminal region of STX17. Consistent with this finding, mature autophagosomes are more negatively charged compared with unclosed intermediate structures. This electrostatic maturation of autophagosomes is likely driven by the accumulation of phosphatidylinositol 4-phosphate (PI4P) in the autophagosomal membrane. Accordingly, dephosphorylation of autophagosomal PI4P prevents the association of STX17 to autophagosomes. Furthermore, molecular dynamics simulations support PI4P-dependent membrane insertion of the transmembrane helices of STX17. Based on these findings, we propose a model in which STX17 recruitment to mature autophagosomes is temporally regulated by a PI4P-driven change in the surface charge of autophagosomes.


Subject(s)
Autophagosomes , Phosphatidylinositol Phosphates , Qa-SNARE Proteins , Qa-SNARE Proteins/metabolism , Qa-SNARE Proteins/genetics , Autophagosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Humans , Molecular Dynamics Simulation , Autophagy/physiology
2.
Proc Natl Acad Sci U S A ; 121(23): e2217971121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38805272

ABSTRACT

Myogenesis is a multistep process that requires a spatiotemporal regulation of cell events resulting finally in myoblast fusion into multinucleated myotubes. Most major insights into the mechanisms underlying fusion seem to be conserved from insects to mammals and include the formation of podosome-like protrusions (PLPs) that exert a driving force toward the founder cell. However, the machinery that governs this process remains poorly understood. In this study, we demonstrate that MTM1 is the main enzyme responsible for the production of phosphatidylinositol 5-phosphate, which in turn fuels PI5P 4-kinase α to produce a minor and functional pool of phosphatidylinositol 4,5-bisphosphate that concentrates in PLPs containing the scaffolding protein Tks5, Dynamin-2, and the fusogenic protein Myomaker. Collectively, our data reveal a functional crosstalk between a PI-phosphatase and a PI-kinase in the regulation of PLP formation.


Subject(s)
Cell Fusion , Myoblasts , Phosphatidylinositol Phosphates , Podosomes , Animals , Phosphatidylinositol Phosphates/metabolism , Mice , Myoblasts/metabolism , Myoblasts/cytology , Podosomes/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Muscle Development/physiology
3.
Virulence ; 15(1): 2350893, 2024 12.
Article in English | MEDLINE | ID: mdl-38725096

ABSTRACT

Coxiella burnetii (C. burnetii) is the causative agent of Q fever, a zoonotic disease. Intracellular replication of C. burnetii requires the maturation of a phagolysosome-like compartment known as the replication permissive Coxiella-containing vacuole (CCV). Effector proteins secreted by the Dot/Icm secretion system are indispensable for maturation of a single large CCV by facilitating the fusion of promiscuous vesicles. However, the mechanisms of CCV maintenance and evasion of host cell clearance remain to be defined. Here, we show that C. burnetii secreted Coxiella vacuolar protein E (CvpE) contributes to CCV biogenesis by inducing lysosome-like vacuole (LLV) enlargement. LLV fission by tubulation and autolysosome degradation is impaired in CvpE-expressing cells. Subsequently, we found that CvpE suppresses lysosomal Ca2+ channel transient receptor potential channel mucolipin 1 (TRPML1) activity in an indirect manner, in which CvpE binds phosphatidylinositol 3-phosphate [PI(3)P] and perturbs PIKfyve activity in lysosomes. Finally, the agonist of TRPML1, ML-SA5, inhibits CCV biogenesis and C. burnetii replication. These results provide insight into the mechanisms of CCV maintenance by CvpE and suggest that the agonist of TRPML1 can be a novel potential treatment that does not rely on antibiotics for Q fever by enhancing Coxiella-containing vacuoles (CCVs) fission.


Subject(s)
Bacterial Proteins , Coxiella burnetii , Lysosomes , Phosphatidylinositol 3-Kinases , Phosphatidylinositol Phosphates , Transient Receptor Potential Channels , Vacuoles , Coxiella burnetii/metabolism , Coxiella burnetii/growth & development , Coxiella burnetii/genetics , Vacuoles/microbiology , Vacuoles/metabolism , Lysosomes/metabolism , Lysosomes/microbiology , Phosphatidylinositol Phosphates/metabolism , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/genetics , Phosphatidylinositol 3-Kinases/metabolism , Animals , Q Fever/microbiology , HeLa Cells , Host-Pathogen Interactions
4.
Sci Signal ; 17(838): eadp3504, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38805585

ABSTRACT

The Hippo pathway blocks epithelial-mesenchymal transition and metastasis in cancer mediated by the transcriptional coactivator YAP. In this issue of Science Signaling, Palamiuc et al. demonstrate that phosphatidylinositol 5-phosphate (PI5P) enhances Hippo pathway activation and that simultaneously the Hippo pathway initiates a positive feedback loop by inhibiting the conversion of PI5P into PIP2.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms , Phosphatidylinositol Phosphates , Protein Serine-Threonine Kinases , Signal Transduction , Humans , Phosphatidylinositol Phosphates/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , Animals , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Hippo Signaling Pathway , Transcription Factors/metabolism , Transcription Factors/genetics
5.
Bioessays ; 46(6): e2400038, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38724256

ABSTRACT

Autophagy, an essential cellular process for maintaining cellular homeostasis and eliminating harmful cytoplasmic objects, involves the de novo formation of double-membraned autophagosomes that engulf and degrade cellular debris, protein aggregates, damaged organelles, and pathogens. Central to this process is the phagophore, which forms from donor membranes rich in lipids synthesized at various cellular sites, including the endoplasmic reticulum (ER), which has emerged as a primary source. The ER-associated omegasomes, characterized by their distinctive omega-shaped structure and accumulation of phosphatidylinositol 3-phosphate (PI3P), play a pivotal role in autophagosome formation. Omegasomes are thought to serve as platforms for phagophore assembly by recruiting essential proteins such as DFCP1/ZFYVE1 and facilitating lipid transfer to expand the phagophore. Despite the critical importance of phagophore biogenesis, many aspects remain poorly understood, particularly the complete range of proteins involved in omegasome dynamics, and the detailed mechanisms of lipid transfer and membrane contact site formation.


Subject(s)
Autophagosomes , Autophagy , Endoplasmic Reticulum , Phosphatidylinositol Phosphates , Autophagosomes/metabolism , Endoplasmic Reticulum/metabolism , Humans , Animals , Phosphatidylinositol Phosphates/metabolism
6.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38781029

ABSTRACT

The mitochondria-ER-cortex anchor (MECA) forms a tripartite membrane contact site between mitochondria, the endoplasmic reticulum (ER), and the plasma membrane (PM). The core component of MECA, Num1, interacts with the PM and mitochondria via two distinct lipid-binding domains; however, the molecular mechanism by which Num1 interacts with the ER is unclear. Here, we demonstrate that Num1 contains a FFAT motif in its C-terminus that interacts with the integral ER membrane protein Scs2. While dispensable for Num1's functions in mitochondrial tethering and dynein anchoring, the FFAT motif is required for Num1's role in promoting mitochondrial division. Unexpectedly, we also reveal a novel function of MECA in regulating the distribution of phosphatidylinositol-4-phosphate (PI(4)P). Breaking Num1 association with any of the three membranes it tethers results in an accumulation of PI(4)P on the PM, likely via disrupting Sac1-mediated PI(4)P turnover. This work establishes MECA as an important regulatory hub that spatially organizes mitochondria, ER, and PM to coordinate crucial cellular functions.


Subject(s)
Endoplasmic Reticulum , Mitochondria , Phosphatidylinositol Phosphates , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Cell Membrane/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mitochondria/metabolism , Mitochondria/genetics , Mitochondrial Dynamics , Phosphatidylinositol Phosphates/metabolism , Protein Binding , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics
7.
Cell Rep ; 43(5): 114119, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38630589

ABSTRACT

Phosphatidylinositol 3-kinase α (PI3Kα) is a heterodimer of p110α catalytic and p85 adaptor subunits that is activated by agonist-stimulated receptor tyrosine kinases. Although p85α recruits p110α to activated receptors on membranes, p85α loss, which occurs commonly in cancer, paradoxically promotes agonist-stimulated PI3K/Akt signaling. p110α localizes to microtubules via microtubule-associated protein 4 (MAP4), facilitating its interaction with activated receptor kinases on endosomes to initiate PI3K/Akt signaling. Here, we demonstrate that in response to agonist stimulation and p85α knockdown, the residual p110α, coupled predominantly to p85ß, exhibits enhanced recruitment with receptor tyrosine kinases to endosomes. Moreover, the p110α C2 domain binds PI3-phosphate, and this interaction is also required to recruit p110α to endosomes and for PI3K/Akt signaling. Stable knockdown of p85α, which mimics the reduced p85α levels observed in cancer, enhances cell growth and tumorsphere formation, and these effects are abrogated by MAP4 or p85ß knockdown, underscoring their role in the tumor-promoting activity of p85α loss.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase , Endosomes , Microtubule-Associated Proteins , Phosphatidylinositol Phosphates , Signal Transduction , Endosomes/metabolism , Humans , Phosphatidylinositol Phosphates/metabolism , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Enzyme Activation , Phosphatidylinositol 3-Kinases/metabolism , Cell Proliferation , Protein Binding
8.
EMBO J ; 43(9): 1740-1769, 2024 May.
Article in English | MEDLINE | ID: mdl-38565949

ABSTRACT

The Hippo pathway effectors Yes-associated protein 1 (YAP) and its homolog TAZ are transcriptional coactivators that control gene expression by binding to TEA domain (TEAD) family transcription factors. The YAP/TAZ-TEAD complex is a key regulator of cancer-specific transcriptional programs, which promote tumor progression in diverse types of cancer, including breast cancer. Despite intensive efforts, the YAP/TAZ-TEAD complex in cancer has remained largely undruggable due to an incomplete mechanistic understanding. Here, we report that nuclear phosphoinositides function as cofactors that mediate the binding of YAP/TAZ to TEADs. The enzymatic products of phosphoinositide kinases PIPKIα and IPMK, including phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and phosphatidylinositol 3,4,5-trisphosphate (P(I3,4,5)P3), bridge the binding of YAP/TAZ to TEAD. Inhibiting these kinases or the association of YAP/TAZ with PI(4,5)P2 and PI(3,4,5)P3 attenuates YAP/TAZ interaction with the TEADs, the expression of YAP/TAZ target genes, and breast cancer cell motility. Although we could not conclusively exclude the possibility that other enzymatic products of IPMK such as inositol phosphates play a role in the mechanism, our results point to a previously unrecognized role of nuclear phosphoinositide signaling in control of YAP/TAZ activity and implicate this pathway as a potential therapeutic target in YAP/TAZ-driven breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing , Breast Neoplasms , Signal Transduction , Trans-Activators , Transcription Factors , YAP-Signaling Proteins , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Transcription Factors/metabolism , Transcription Factors/genetics , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Female , Trans-Activators/metabolism , Trans-Activators/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Cell Line, Tumor , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositols/metabolism , Gene Expression Regulation, Neoplastic , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cell Nucleus/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics
9.
Biochemistry ; 63(9): 1097-1106, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38669178

ABSTRACT

As a key component for NADPH oxidase 2 (NOX2) activation, the peripheral membrane protein p47phox translocates a cytosolic activating complex to the membrane through its PX domain. This study elucidates a potential regulatory mechanism of p47phox recruitment and NOX2 activation by inositol hexaphosphate (IP6). Through NMR, fluorescence polarization, and FRET experimental results, IP6 is shown to be capable of breaking the lipid binding and membrane anchoring events of p47phox-PX with low micromolar potency. Other phosphorylated inositol species such as IP5(1,3,4,5,6), IP4(1,3,4,5), and IP3(1,3,4) show weaker binding and no ability to inhibit lipid interactions in physiological concentration ranges. The low micromolar potency of IP6 inhibition of the p47phox membrane anchoring suggests that physiologically relevant concentrations of IP6 serve as regulators, as seen in other membrane anchoring domains. The PX domain of p47phox is known to be promiscuous to a variety of phosphatidylinositol phosphate (PIP) lipids, and this regulation may help target the domain only to the membranes most highly enriched with the highest affinity PIPs, such as the phagosomal membrane, while preventing aberrant binding to other membranes with high and heterogeneous PIP content, such as the plasma membrane. This study provides insight into a potential novel regulatory mechanism behind NOX2 activation and reveals a role for small-molecule regulation in this important NOX2 activator.


Subject(s)
NADPH Oxidases , Phytic Acid , Phytic Acid/metabolism , Phytic Acid/chemistry , NADPH Oxidases/metabolism , NADPH Oxidases/antagonists & inhibitors , Humans , Cell Membrane/metabolism , NADPH Oxidase 2/metabolism , Phosphatidylinositol Phosphates/metabolism
10.
J Cell Biol ; 223(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38578646

ABSTRACT

Phosphoinositides are a small family of phospholipids that act as signaling hubs and key regulators of cellular function. Detecting their subcellular distribution is crucial to gain insights into membrane organization and is commonly done by the overexpression of biosensors. However, this leads to cellular perturbations and is challenging in systems that cannot be transfected. Here, we present a toolkit for the reliable, fast, multiplex, and super-resolution detection of phosphoinositides in fixed cells and tissue, based on recombinant biosensors with self-labeling SNAP tags. These are highly specific and reliably visualize the subcellular distributions of phosphoinositides across scales, from 2D or 3D cell culture to Drosophila tissue. Further, these probes enable super-resolution approaches, and using STED microscopy, we reveal the nanoscale organization of PI(3)P on endosomes and PI(4)P on the Golgi. Finally, multiplex staining reveals an unexpected presence of PI(3,5)P2-positive membranes in swollen lysosomes following PIKfyve inhibition. This approach enables the versatile, high-resolution visualization of multiple phosphoinositide species in an unprecedented manner.


Subject(s)
Biosensing Techniques , Phosphatidylinositols , Endosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Biosensing Techniques/methods
11.
Dev Cell ; 59(7): 911-923.e4, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38447569

ABSTRACT

Autophagy eliminates cytoplasmic material by engulfment in membranous vesicles targeted for lysosome degradation. Nonselective autophagy coordinates sequestration of bulk cargo with the growth of the isolation membrane (IM) in a yet-unknown manner. Here, we show that in the budding yeast Saccharomyces cerevisiae, IMs expand while maintaining a rim sufficiently wide for sequestration of large cargo but tight enough to mature in due time. An obligate complex of Atg24/Snx4 with Atg20 or Snx41 assembles locally at the rim in a spatially extended manner that specifically depends on autophagic PI(3)P. This assembly stabilizes the open rim to promote autophagic sequestration of large cargo in correlation with vesicle expansion. Moreover, constriction of the rim by the PI(3)P-dependent Atg2-Atg18 complex and clearance of PI(3)P by Ymr1 antagonize rim opening to promote autophagic maturation and consumption of small cargo. Tight regulation of membrane rim aperture by PI(3)P thus couples the mechanism and physiology of nonselective autophagy.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Autophagy/physiology , Phosphatidylinositol Phosphates/metabolism , Autophagy-Related Proteins/metabolism , Autophagosomes/metabolism
12.
Cell Rep ; 43(4): 113992, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38536815

ABSTRACT

Insulin is packaged into secretory granules that depart the Golgi and undergo a maturation process that involves changes in the protein and lipid composition of the granules. Here, we show that insulin secretory granules form physical contacts with the endoplasmic reticulum and that the lipid exchange protein oxysterol-binding protein (OSBP) is recruited to these sites in a Ca2+-dependent manner. OSBP binding to insulin granules is positively regulated by phosphatidylinositol-4 (PI4)-kinases and negatively regulated by the PI4 phosphate (PI(4)P) phosphatase Sac2. Loss of Sac2 results in excess accumulation of cholesterol on insulin granules that is normalized when OSBP expression is reduced, and both acute inhibition and small interfering RNA (siRNA)-mediated knockdown of OSBP suppress glucose-stimulated insulin secretion without affecting insulin production or intracellular Ca2+ signaling. In conclusion, we show that lipid exchange at endoplasmic reticulum (ER)-granule contact sites is involved in the exocytic process and propose that these contacts act as reaction centers with multimodal functions during insulin granule maturation.


Subject(s)
Cholesterol , Endoplasmic Reticulum , Insulin Secretion , Insulin , Minor Histocompatibility Antigens , Receptors, Steroid , Secretory Vesicles , Endoplasmic Reticulum/metabolism , Secretory Vesicles/metabolism , Animals , Cholesterol/metabolism , Insulin/metabolism , Receptors, Steroid/metabolism , Phosphatidylinositol Phosphates/metabolism , Mice , Humans , Calcium/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Glucose/metabolism
13.
Biophys J ; 123(9): 1058-1068, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38515298

ABSTRACT

Phosphatidylinositol (3,4,5)-trisphosphate (PIP3) is a signaling lipid on the plasma membrane that plays a fundamental role in cell signaling with a strong impact on cell physiology and diseases. It is responsible for the protruding edge formation, cell polarization, macropinocytosis, and other membrane remodeling dynamics in cells. It has been shown that the membrane confinement and curvature affects the wave formation of PIP3 and F-actin. But, even in the absence of F-actin, a complex self-organization of the spatiotemporal PIP3 waves is observed. In recent findings, we have shown that these waves can be guided and pinned on strongly bended Dictyostelium membranes caused by molecular crowding and curvature-limited diffusion. Based on these experimental findings, we investigate the spatiotemporal PIP3 wave dynamics on realistic three-dimensional cell-like membranes to explore the effect of curvature-limited diffusion, as observed experimentally. We use an established stochastic reaction-diffusion model with enzymatic Michaelis-Menten-type reactions that mimics the dynamics of Dictyostelium cells. As these cells mimic the three-dimensional shape and size observed experimentally, we found that the PIP3 wave directionality can be explained by a Hopf-like and a reverse periodic-doubling bifurcation for uniform diffusion and curvature-limited diffusion properties. Finally, we compare the results with recent experimental findings and discuss the discrepancy between the biological and numerical results.


Subject(s)
Cell Membrane , Dictyostelium , Models, Biological , Phosphatidylinositol Phosphates , Cell Membrane/metabolism , Dictyostelium/cytology , Dictyostelium/metabolism , Phosphatidylinositol Phosphates/metabolism , Diffusion
14.
J Clin Lab Anal ; 38(7): e25031, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38514901

ABSTRACT

BACKGROUND: Primary cilia are static microtubule-based structures protruding from the cell surface and present on most vertebrate cells. The appropriate localization of phospholipids is essential for cilia formation and stability. INPP5E is a cilia-localized inositol 5-phosphatase; its deletion alters the phosphoinositide composition in the ciliary membrane, disrupting ciliary function. METHODS: The EGFP-2xP4MSidM, PHPLCδ1-EGFP, and SMO-tRFP plasmids were constructed by the Gateway system to establish a stable RPE1 cell line. The INPP5E KO RPE1 cell line was constructed with the CRISPR/Cas9 system. The localization of INPP5E and the distribution of PI(4,5)P2 and PI4P were examined by immunofluorescence microscopy. The fluorescence intensity co-localized with cilia was quantified by ImageJ. RESULTS: In RPE1 cells, PI4P is localized at the ciliary membrane, whereas PI(4,5)P2 is localized at the base of cilia. Knocking down or knocking out INPP5E alters this distribution, resulting in the distribution of PI(4,5)P2 along the ciliary membrane and the disappearance of PI4P from the cilia. Meanwhile, PI(4,5)P2 is located in the ciliary membrane labeled by SMO-tRFP. CONCLUSIONS: INPP5E regulates the distribution of phosphoinositide on cilia. PI(4,5)P2 localizes at the ciliary membrane labeled with SMO-tRFP, indicating that ciliary pocket membrane contains PI(4,5)P2, and phosphoinositide composition in early membrane structures may differ from that in mature ciliary membrane.


Subject(s)
Cilia , Phosphoric Monoester Hydrolases , Cilia/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/genetics , Humans , Cell Line , Phosphatidylinositol 4,5-Diphosphate/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/cytology , Phosphatidylinositol Phosphates/metabolism , CRISPR-Cas Systems , Phospholipids/metabolism
15.
Proc Natl Acad Sci U S A ; 121(10): e2315493121, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38408242

ABSTRACT

Oxysterol-binding protein-related proteins (ORPs) play key roles in the distribution of lipids in eukaryotic cells by exchanging sterol or phosphatidylserine for PI4P between the endoplasmic reticulum (ER) and other cell regions. However, it is unclear how their exchange capacity is coupled to PI4P metabolism. To address this question quantitatively, we analyze the activity of a representative ORP, Osh4p, in an ER/Golgi interface reconstituted with ER- and Golgi-mimetic membranes functionalized with PI4P phosphatase Sac1p and phosphatidylinositol (PI) 4-kinase, respectively. Using real-time assays, we demonstrate that upon adenosine triphosphate (ATP) addition, Osh4p creates a sterol gradient between these membranes, relying on the spatially distant synthesis and hydrolysis of PI4P, and quantify how much PI4P is needed for this process. Then, we develop a quantitatively accurate kinetic model, validated by our data, and extrapolate this to estimate to what extent PI4P metabolism can drive ORP-mediated sterol transfer in cells. Finally, we show that Sec14p can support PI4P metabolism and Osh4p activity by transferring PI between membranes. This study establishes that PI4P synthesis drives ORP-mediated lipid exchange and that ATP energy is needed to generate intermembrane lipid gradients. Furthermore, it defines to what extent ORPs can distribute lipids in the cell and reassesses the role of PI-transfer proteins in PI4P metabolism.


Subject(s)
Phosphatidylinositol Phosphates , Receptors, Steroid , Phosphatidylinositol Phosphates/metabolism , Biological Transport , Sterols/metabolism , Phosphatidylserines/metabolism , Lipid Metabolism , Adenosine Triphosphate/metabolism , Cell Membrane/metabolism , Receptors, Steroid/metabolism
16.
STAR Protoc ; 5(1): 102759, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38088931

ABSTRACT

Phosphatidylinositol 3-phosphate (PI3P) levels govern membrane trafficking in Candida glabrata. Here, we present a confocal imaging-based protocol for PI3P localization analysis using the GFP-FYVE (found in Fab1, YOTB, Vac1, and EEA1) fusion protein. We describe steps for cloning the FYVE domain into the GFP-containing vector backbone, transforming FYVE-GFP into C. glabrata, and preparing slides with FYVE-GFP-expressing C. glabrata cells. We then detail procedures for acquiring and analyzing images and quantifying signal data. This protocol is adaptable to subcellular localization analysis of other low-abundant lipid and protein molecules. For complete details on the use and execution of this protocol, please refer to Askari et al. (2023).1.


Subject(s)
Candida glabrata , Phosphatidylinositol Phosphates , Candida glabrata/genetics , Candida glabrata/metabolism , Amino Acid Sequence , Phosphatidylinositol Phosphates/metabolism , Microscopy, Confocal
17.
Plant J ; 117(1): 212-225, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37828913

ABSTRACT

Phosphatidylinositol 4-phosphate 5-kinase (PIP5K) is a key enzyme producing the signaling lipid phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2 ] in eukaryotes. Although PIP5K genes are reported to be involved in pollen tube germination and growth, the essential roles of PIP5K in these processes remain unclear. Here, we performed a comprehensive genetic analysis of the Arabidopsis thaliana PIP5K4, PIP5K5, and PIP5K6 genes and revealed that their redundant function is essential for pollen germination. Pollen with the pip5k4pip5k5pip5k6 triple mutation was sterile, while pollen germination efficiency and pollen tube growth were reduced in the pip5k6 single mutant and further reduced in the pip5k4pip5k6 and pip5k5pip5k6 double mutants. YFP-fusion proteins, PIP5K4-YFP, PIP5K5-YFP, and PIP5K6-YFP, which could rescue the sterility of the triple mutant pollen, preferentially localized to the tricolpate aperture area and the future germination site on the plasma membrane prior to germination. Triple mutant pollen grains under the germination condition, in which spatiotemporal localization of the PtdIns(4,5)P2 fluorescent marker protein 2xmCHERRY-2xPHPLC as seen in the wild type was abolished, exhibited swelling and rupture of the pollen wall, but neither the conspicuous protruding site nor site-specific deposition of cell wall materials for germination. These data indicate that PIP5K4-6 and their product PtdIns(4,5)P2 are essential for pollen germination, possibly through the establishment of the germination polarity in a pollen grain.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Germination/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Pollen Tube/metabolism , Pollen
18.
J Biol Chem ; 299(12): 105473, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37979916

ABSTRACT

Vacuolar H+-ATPases (V-ATPases) are highly conserved multisubunit enzymes that maintain the distinct pH of eukaryotic organelles. The integral membrane a-subunit is encoded by tissue- and organelle-specific isoforms, and its cytosolic N-terminal domain (aNT) modulates organelle-specific regulation and targeting of V-ATPases. Organelle membranes have specific phosphatidylinositol phosphate (PIP) lipid enrichment linked to maintenance of organelle pH. In yeast, the aNT domains of the two a-subunit isoforms bind PIP lipids enriched in the organelle membranes where they reside; these interactions affect activity and regulatory properties of the V-ATPases containing each isoform. Humans have four a-subunit isoforms, and we hypothesize that the aNT domains of these isoforms will also bind to specific PIP lipids. The a1 and a2 isoforms of human V-ATPase a-subunits are localized to endolysosomes and Golgi, respectively. We determined that bacterially expressed Hua1NT and Hua2NT bind specifically to endolysosomal PIP lipids PI(3)P and PI(3,5)P2 and Golgi enriched PI(4)P, respectively. Despite the lack of canonical PIP-binding sites, we identified potential binding sites in the HuaNT domains by sequence comparisons and existing subunit structures and models. We found that mutations at a similar location in the distal loops of both HuaNT isoforms compromise binding to their cognate PIP lipids, suggesting that these loops encode PIP specificity of the a-subunit isoforms. These data suggest a mechanism through which PIP lipid binding could stabilize and activate V-ATPases in distinct organelles.


Subject(s)
Phosphatidylinositol Phosphates , Protein Subunits , Vacuolar Proton-Translocating ATPases , Humans , Binding Sites , Endosomes/enzymology , Endosomes/metabolism , Golgi Apparatus/enzymology , Golgi Apparatus/metabolism , Hydrogen-Ion Concentration , Lysosomes/enzymology , Lysosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Substrate Specificity , Vacuolar Proton-Translocating ATPases/chemistry , Vacuolar Proton-Translocating ATPases/metabolism , Protein Domains
19.
Commun Biol ; 6(1): 1008, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37794132

ABSTRACT

Phagosome maturation is critical for immune defense, defining whether ingested material is destroyed or converted into antigens. Sec22b regulates phagosome maturation, yet how has remained unclear. Here we show Sec22b tethers endoplasmic reticulum-phagosome membrane contact sites (MCS) independently of the known tether STIM1. Sec22b knockdown increases calcium signaling, phagolysosome fusion and antigen degradation and alters phagosomal phospholipids PI(3)P, PS and PI(4)P. Levels of PI(4)P, a lysosome docking lipid, are rescued by Sec22b re-expression and by expression of the artificial tether MAPPER but not the MCS-disrupting mutant Sec22b-P33. Moreover, Sec22b co-precipitates with the PS/PI(4)P exchange protein ORP8. Wild-type, but not mutant ORP8 rescues phagosomal PI(4)P and reduces antigen degradation. Sec22b, MAPPER and ORP8 but not P33 or mutant-ORP8 restores phagolysosome fusion in knockdown cells. These findings clarify an alternative mechanism through which Sec22b controls phagosome maturation and beg a reassessment of the relative contribution of Sec22b-mediated fusion versus tethering to phagosome biology.


Subject(s)
Phagocytosis , Phagosomes , Phagosomes/metabolism , Phagocytosis/physiology , Endoplasmic Reticulum/metabolism , Phosphatidylinositol Phosphates/metabolism
20.
Science ; 382(6667): 155-156, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824634

ABSTRACT

Phosphatidylinositol 3,5-bisphosphate enables transport of proteins to synaptic sites.


Subject(s)
Phosphatidylinositol Phosphates , Signal Transduction , Synapses , Animals , Humans , Mice , Neurogenesis , Protein Transport , Synapses/metabolism , Phosphatidylinositol Phosphates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...