Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96.165
Filter
1.
Methods Mol Biol ; 2814: 45-53, 2024.
Article in English | MEDLINE | ID: mdl-38954196

ABSTRACT

Eukaryotic cells have been constantly challenged throughout their evolution by pathogens, mechanical stresses, or toxic compounds that induce plasma membrane (PM) or endolysosomal membrane damage. The survival of the wounded cells depends on damage detection and repair machineries that are evolutionary conserved between protozoan, plants, and animals. We use the social amoeba Dictyostelium discoideum as a model system to study bacteria, mechanical or sterile membrane damage that allows us to identify and monitor factors involved in PM, endolysosomal damage response (ELDR), and endolysosomal homeostasis. Importantly, the sterile damage techniques presented here homogenously affect cell populations, which allows to phenotype mutant strains and quantify various aspects of cell fitness using live cell microscopy. This is instrumental to functionally assess genes involved in the repair of damaged plasma membrane or intracellular compartments and the degradation of extensively damaged compartments. Here, we describe how to inflict sterile PM or endolysosomal membrane damage, how to monitor the cell-intrinsic response to damage, and how to proxy proton leakage from damaged acidic compartments and quantify cell viability.


Subject(s)
Cell Membrane , Dictyostelium , Lysosomes , Dictyostelium/genetics , Dictyostelium/metabolism , Cell Membrane/metabolism , Lysosomes/metabolism , Cell Survival
2.
Methods Mol Biol ; 2814: 133-147, 2024.
Article in English | MEDLINE | ID: mdl-38954203

ABSTRACT

Activation processes at the plasma membrane have been studied with life-cell imaging using GFP fused to a protein that binds to a component of the activation process. In this way, PIP3 formation has been monitored with CRAC-GFP, Ras-GTP with RBD-Raf-GFP, and Rap-GTP with Ral-GDS-GFP. The fluorescent sensors translocate from the cytoplasm to the plasma membrane upon activation of the process. Although this translocation assay can provide very impressive images and movies, the method is not very sensitive, and amount of GFP-sensor at the plasma membrane is not linear with the amount of activator. The fluorescence in pixels at the cell boundary is partly coming from the GFP-sensor that is bound to the activated membrane and partly from unbound GFP-sensor in the cytosolic volume of that boundary pixel. The variable and unknown amount of cytosol in boundary pixels causes the low sensitivity and nonlinearity of the GFP-translocation assay. Here we describe a method in which the GFP-sensor is co-expressed with cytosolic-RFP. For each boundary pixels, the RFP fluorescence is used to determine the amount of cytosol of that pixel and is subtracted from the GFP fluorescence of that pixel yielding the amount of GFP-sensor that is specifically associated with the plasma membrane in that pixel. This GRminusRD method using GFP-sensor/RFP is at least tenfold more sensitive, more reproducible, and linear with activator compared to GFP-sensor alone.


Subject(s)
Cell Membrane , Green Fluorescent Proteins , Cell Membrane/metabolism , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Humans , Luminescent Proteins/metabolism , Luminescent Proteins/genetics , Protein Transport , Microscopy, Fluorescence/methods , Cytosol/metabolism , Animals
3.
PLoS One ; 19(7): e0306142, 2024.
Article in English | MEDLINE | ID: mdl-38954698

ABSTRACT

Developing T1-weighted magnetic resonance imaging (MRI) contrast agents with enhanced biocompatibility and targeting capabilities is crucial owing to concerns over current agents' potential toxicity and suboptimal performance. Drawing inspiration from "biomimetic camouflage," we isolated cell membranes (CMs) from human glioblastoma (T98G) cell lines via the extrusion method to facilitate homotypic glioma targeting. At an 8:1 mass ratio of ferric chloride hexahydrate to gallic acid (GA), the resulting iron (Fe)-GA nanoparticles (NPs) proved effective as a T1-weighted MRI contrast agent. T98G CM-coated Fe-GA NPs demonstrated improved homotypic glioma targeting, validated through Prussian blue staining and in vitro MRI. This biomimetic camouflage strategy holds promise for the development of targeted theranostic agents in a safe and effective manner.


Subject(s)
Contrast Media , Gallic Acid , Magnetic Resonance Imaging , Gallic Acid/chemistry , Humans , Magnetic Resonance Imaging/methods , Cell Line, Tumor , Contrast Media/chemistry , Iron/chemistry , Biomimetic Materials/chemistry , Glioblastoma/drug therapy , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Nanoparticles/chemistry , Ferric Compounds/chemistry , Cell Membrane/metabolism
4.
Commun Biol ; 7(1): 791, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951588

ABSTRACT

According to single-molecule localisation microscopy almost all plasma membrane proteins are clustered. We demonstrate that clusters can arise from variations in membrane topography where the local density of a randomly distributed membrane molecule to a degree matches the variations in the local amount of membrane. Further, we demonstrate that this false clustering can be differentiated from genuine clustering by using a membrane marker to report on local variations in the amount of membrane. In dual colour live cell single molecule localisation microscopy using the membrane probe DiI alongside either the transferrin receptor or the GPI-anchored protein CD59, we found that pair correlation analysis reported both proteins and DiI as being clustered, as did its derivative pair correlation-photoactivation localisation microscopy and nearest neighbour analyses. After converting the localisations into images and using the DiI image to factor out topography variations, no CD59 clusters were visible, suggesting that the clustering reported by the other methods is an artefact. However, the TfR clusters persisted after topography variations were factored out. We demonstrate that membrane topography variations can make membrane molecules appear clustered and present a straightforward remedy suitable as the first step in the cluster analysis pipeline.


Subject(s)
CD59 Antigens , Cell Membrane , Receptors, Transferrin , Single Molecule Imaging , Single Molecule Imaging/methods , Cell Membrane/metabolism , CD59 Antigens/metabolism , Receptors, Transferrin/metabolism , Humans , Membrane Proteins/metabolism , Cluster Analysis , Microscopy, Fluorescence/methods
5.
Commun Biol ; 7(1): 795, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951640

ABSTRACT

The peroxisome is a versatile organelle that performs diverse metabolic functions. PEX3, a critical regulator of the peroxisome, participates in various biological processes associated with the peroxisome. Whether PEX3 is involved in peroxisome-related redox homeostasis and myocardial regenerative repair remains elusive. We investigate that cardiomyocyte-specific PEX3 knockout (Pex3-KO) results in an imbalance of redox homeostasis and disrupts the endogenous proliferation/development at different times and spatial locations. Using Pex3-KO mice and myocardium-targeted intervention approaches, the effects of PEX3 on myocardial regenerative repair during both physiological and pathological stages are explored. Mechanistically, lipid metabolomics reveals that PEX3 promotes myocardial regenerative repair by affecting plasmalogen metabolism. Further, we find that PEX3-regulated plasmalogen activates the AKT/GSK3ß signaling pathway via the plasma membrane localization of ITGB3. Our study indicates that PEX3 may represent a novel therapeutic target for myocardial regenerative repair following injury.


Subject(s)
Cell Membrane , Integrin beta3 , Mice, Knockout , Regeneration , Animals , Mice , Integrin beta3/metabolism , Integrin beta3/genetics , Cell Membrane/metabolism , Myocytes, Cardiac/metabolism , Male , Plasmalogens/metabolism , Signal Transduction , Myocardium/metabolism , Myocardium/pathology , Mice, Inbred C57BL , Heart Injuries/metabolism , Heart Injuries/pathology , Heart Injuries/genetics , Cell Proliferation , Membrane Proteins/metabolism , Membrane Proteins/genetics
6.
Theranostics ; 14(9): 3423-3438, 2024.
Article in English | MEDLINE | ID: mdl-38948056

ABSTRACT

PRL1 and PRL3, members of the protein tyrosine phosphatase family, have been associated with cancer metastasis and poor prognosis. Despite extensive research on their protein phosphatase activity, their potential role as lipid phosphatases remains elusive. Methods: We conducted comprehensive investigations to elucidate the lipid phosphatase activity of PRL1 and PRL3 using a combination of cellular assays, biochemical analyses, and protein interactome profiling. Functional studies were performed to delineate the impact of PRL1/3 on macropinocytosis and its implications in cancer biology. Results: Our study has identified PRL1 and PRL3 as lipid phosphatases that interact with phosphoinositide (PIP) lipids, converting PI(3,4)P2 and PI(3,5)P2 into PI(3)P on the cellular membranes. These enzymatic activities of PRLs promote the formation of membrane ruffles, membrane blebbing and subsequent macropinocytosis, facilitating nutrient extraction, cell migration, and invasion, thereby contributing to tumor development. These enzymatic activities of PRLs promote the formation of membrane ruffles, membrane blebbing and subsequent macropinocytosis. Additionally, we found a correlation between PRL1/3 expression and glioma development, suggesting their involvement in glioma progression. Conclusions: Combining with the knowledge that PRLs have been identified to be involved in mTOR, EGFR and autophagy, here we concluded the physiological role of PRL1/3 in orchestrating the nutrient sensing, absorbing and recycling via regulating macropinocytosis through its lipid phosphatase activity. This mechanism could be exploited by tumor cells facing a nutrient-depleted microenvironment, highlighting the potential therapeutic significance of targeting PRL1/3-mediated macropinocytosis in cancer treatment.


Subject(s)
Pinocytosis , Protein Tyrosine Phosphatases , Protein Tyrosine Phosphatases/metabolism , Humans , Cell Line, Tumor , Animals , Neoplasm Proteins/metabolism , Cell Movement , Mice , Cell Membrane/metabolism , Phosphatidylinositols/metabolism , Membrane Proteins , Cell Cycle Proteins
7.
PLoS One ; 19(6): e0304172, 2024.
Article in English | MEDLINE | ID: mdl-38861534

ABSTRACT

There is a close relation between membrane receptor dynamics and their behavior. Several microscopy techniques have been developed to study protein dynamics in live cells such as the Fluorescence Recovery After Photobleaching (FRAP) or the Single Particle Tracking (SPT). These methodologies require expensive instruments, are time consuming, allow the analysis of small portion of the cell or an extremely small number of receptors at a time. Here we propose a time-saving approach that allows to visualize the entire receptor pool and its localization in time. This protocol requires an epifluorescence microscope equipped for structured illuminated sectioning and for live cell imaging. It can be applied to characterize membrane receptor and multi protein complex and their response to activators or inhibitors. Image acquisition and analysis can be performed in two days, while cells and substratum preparation require a few minutes a day for three days.


Subject(s)
Cell Membrane , Fluorescence Recovery After Photobleaching , Microscopy, Fluorescence , Cell Membrane/metabolism , Humans , Fluorescence Recovery After Photobleaching/methods , Microscopy, Fluorescence/methods , Animals , Receptors, Cell Surface/metabolism
8.
World J Microbiol Biotechnol ; 40(8): 243, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38869625

ABSTRACT

It was known that UVc irradiation increases the reactive oxygen species' (ROS) levels in bacteria hence the intervention of antioxidant enzymes and causes also changes in fatty acids (FAs) composition enabling bacteria to face antibiotics. Here, we intended to elucidate an interrelationship between SOD and susceptibility to antibiotics by studying FA membrane composition of UVc-treated P. aeruginosa PAO1 and its isogenic mutants (sodM, sodB and sod MB) membrane, after treatment with antibiotics. Swarmer mutants defective in genes encoding superoxide dismutase were pre-exposed to UVc radiations and then tested by disk diffusion method for their contribution to antibiotic tolerance in comparison with the P. aeruginosa wild type (WT). Moreover, fatty acid composition of untreated and UVc-treated WT and sod mutants was examined by Gaz chromatography and correlated to antibiotic resistance. Firstly, it has been demonstrated that after UVc exposure, swarmer WT strain, sodM and sodB mutants remain resistant to polymixin B, a membrane target antibiotic, through membrane unsaturation supported by the intervention of Mn-SOD after short UVc exposure and cyclopropanation of unsaturated FAs supported by the action of Fe-SOD after longer UVc exposure. However, resistance for ciprofloxacin is correlated with increase in saturated FAs. This correlation has been confirmed by a molecular docking approach showing that biotin carboxylase, involved in the initial stage of FA biosynthesis, exhibits a high affinity for ciprofloxacin. This investigation has explored the correlation of antibiotic resistance with FA content of swarmer P.aeruginosa pre-exposed to UVc radiations, confirmed to be antibiotic target dependant.


Subject(s)
Anti-Bacterial Agents , Mutation , Pseudomonas aeruginosa , Superoxide Dismutase , Ultraviolet Rays , Anti-Bacterial Agents/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Cyclopropanes/pharmacology , Drug Resistance, Bacterial/genetics , Fatty Acids/metabolism , Ciprofloxacin/pharmacology , Microbial Sensitivity Tests , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Computer Simulation , Polymyxin B/pharmacology
9.
Signal Transduct Target Ther ; 9(1): 158, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38862461

ABSTRACT

Cell membrane-camouflaged nanoparticles possess inherent advantages derived from their membrane structure and surface antigens, including prolonged circulation in the bloodstream, specific cell recognition and targeting capabilities, and potential for immunotherapy. Herein, we introduce a cell membrane biomimetic nanodrug platform termed MPB-3BP@CM NPs. Comprising microporous Prussian blue nanoparticles (MPB NPs) serving as both a photothermal sensitizer and carrier for 3-bromopyruvate (3BP), these nanoparticles are cloaked in a genetically programmable cell membrane displaying variants of signal regulatory protein α (SIRPα) with enhanced affinity to CD47. As a result, MPB-3BP@CM NPs inherit the characteristics of the original cell membrane, exhibiting an extended circulation time in the bloodstream and effectively targeting CD47 on the cytomembrane of colorectal cancer (CRC) cells. Notably, blocking CD47 with MPB-3BP@CM NPs enhances the phagocytosis of CRC cells by macrophages. Additionally, 3BP, an inhibitor of hexokinase II (HK2), suppresses glycolysis, leading to a reduction in adenosine triphosphate (ATP) levels and lactate production. Besides, it promotes the polarization of tumor-associated macrophages (TAMs) towards an anti-tumor M1 phenotype. Furthermore, integration with MPB NPs-mediated photothermal therapy (PTT) enhances the therapeutic efficacy against tumors. These advantages make MPB-3BP@CM NPs an attractive platform for the future development of innovative therapeutic approaches for CRC. Concurrently, it introduces a universal approach for engineering disease-tailored cell membranes for tumor therapy.


Subject(s)
CD47 Antigen , Cell Membrane , Colorectal Neoplasms , Nanoparticles , Colorectal Neoplasms/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Nanoparticles/chemistry , Humans , CD47 Antigen/genetics , Mice , Cell Membrane/metabolism , Cell Membrane/genetics , Animals , Pyruvates/chemistry , Pyruvates/pharmacology , Hexokinase/genetics , Cell Line, Tumor , Macrophages/metabolism , Macrophages/drug effects , Ferrocyanides
10.
Phys Chem Chem Phys ; 26(23): 16529-16539, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38828872

ABSTRACT

This study reports on the effects of conformationally controlled amphiphilicity of antimicrobial peptides (AMPs) on their ability to coat TiO2 nanoparticles (NPs) and boost the photocatalytic antimicrobial effects of such NPs. For this, TiO2 NPs were combined with AMP EFK17 (EFKRIVQRIKDFLRNLV), displaying a disordered conformation in aqueous solution but helix formation on interaction with bacterial membranes. The membrane-bound helix is amphiphilic, with all polar and charged amino acid residues located at one side and all non-polar and hydrophobic residues on the other. In contrast, the d-enantiomer variant EFK17-d (E(dF)KR(dI)VQR(dI)KD(dF)LRNLV) is unable to form the amphiphilic helix on bacterial membrane interaction, whereas the W-residues in EFK17-W (EWKRWVQRWKDFLRNLV) boost hydrophobic interactions of the amphiphilic helix. Circular dichroism results showed the effects displayed for the free peptide, to also be present for peptide-coated TiO2 NPs, causing peptide binding to decrease in the order EFK17-W > EFK17 > EFK17-d. Notably, the formation of reactive oxygen species (ROS) by the TiO2 NPs was essentially unaffected by the presence of peptide coating, for all the peptides investigated, and the coatings stabilized over hours of UV exposure. Photocatalytic membrane degradation from TiO2 NPs coated with EFK17-W and EFK17 was promoted for bacteria-like model bilayers containing anionic phosphatidylglycerol but suppressed in mammalian-like bilayers formed by zwitterionic phosphatidylcholine and cholesterol. Structural aspects of these effects were further investigated by neutron reflectometry with clear variations observed between the bacteria- and mammalian-like model bilayers for the three peptides. Mirroring these results in bacteria-like model membranes, combining TiO2 NPs with EFK17-W and EFK17, but not with non-adsorbing EFK17-d, resulted in boosted antimicrobial effects of the resulting cationic composite NPs already in darkness, effects enhanced further on UV illumination.


Subject(s)
Titanium , Titanium/chemistry , Titanium/pharmacology , Catalysis , Nanoparticles/chemistry , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Reactive Oxygen Species/metabolism , Hydrophobic and Hydrophilic Interactions , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology
11.
Elife ; 122024 Jun 05.
Article in English | MEDLINE | ID: mdl-38837189

ABSTRACT

The organelles of eukaryotic cells maintain distinct protein and lipid compositions required for their specific functions. The mechanisms by which many of these components are sorted to their specific locations remain unknown. While some motifs mediating subcellular protein localization have been identified, many membrane proteins and most membrane lipids lack known sorting determinants. A putative mechanism for sorting of membrane components is based on membrane domains known as lipid rafts, which are laterally segregated nanoscopic assemblies of specific lipids and proteins. To assess the role of such domains in the secretory pathway, we applied a robust tool for synchronized secretory protein traffic (RUSH, Retention Using Selective Hooks) to protein constructs with defined affinity for raft phases. These constructs consist solely of single-pass transmembrane domains (TMDs) and, lacking other sorting determinants, constitute probes for membrane domain-mediated trafficking. We find that while raft affinity can be sufficient for steady-state PM localization, it is not sufficient for rapid exit from the endoplasmic reticulum (ER), which is instead mediated by a short cytosolic peptide motif. In contrast, we find that Golgi exit kinetics are highly dependent on raft affinity, with raft preferring probes exiting the Golgi ~2.5-fold faster than probes with minimal raft affinity. We rationalize these observations with a kinetic model of secretory trafficking, wherein Golgi export can be facilitated by protein association with raft domains. These observations support a role for raft-like membrane domains in the secretory pathway and establish an experimental paradigm for dissecting its underlying machinery.


Subject(s)
Endoplasmic Reticulum , Golgi Apparatus , Membrane Microdomains , Protein Transport , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Membrane Microdomains/metabolism , Secretory Pathway , Humans , Kinetics , Cell Membrane/metabolism , Membrane Proteins/metabolism , HeLa Cells
12.
Bull Math Biol ; 86(8): 87, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874691

ABSTRACT

In the context of protein-protein binding, the dissociation constant is used to describe the affinity between two proteins. For protein-protein interactions, most experimentally-measured dissociation constants are measured in solution and reported in units of volume concentration. However, many protein interactions take place on membranes. These interactions have dissociation constants with units of areal concentration, rather than volume concentration. Here, we present a novel, stochastic approach to understanding the dimensional dependence of binding kinetics. Using stochastic exit time calculations, in discrete and continuous space, we derive general reaction rates for protein-protein binding in one, two, and three dimensions and demonstrate that dimensionality greatly affects binding kinetics. Further, we present a formula to transform three-dimensional experimentally-measured dissociation constants to two-dimensional dissociation constants. This conversion can be used to mathematically model binding events that occur on membranes.


Subject(s)
Mathematical Concepts , Models, Biological , Protein Binding , Stochastic Processes , Kinetics , Cell Membrane/metabolism , Computer Simulation , Proteins/metabolism , Proteins/chemistry
13.
Nat Commun ; 15(1): 4986, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862544

ABSTRACT

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


Subject(s)
Cell Membrane , Focal Adhesions , Talin , Vinculin , Talin/metabolism , Talin/chemistry , Focal Adhesions/metabolism , Cell Membrane/metabolism , Vinculin/metabolism , Vinculin/chemistry , Humans , Animals , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Integrins/metabolism , Integrins/chemistry , Cytoplasm/metabolism , Protein Binding , Phase Separation
14.
Sci Immunol ; 9(96): eadl2388, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38848343

ABSTRACT

Professional phagocytes like neutrophils and macrophages tightly control what they consume, how much they consume, and when they move after cargo uptake. We show that plasma membrane abundance is a key arbiter of these cellular behaviors. Neutrophils and macrophages lacking the G protein subunit Gß4 exhibited profound plasma membrane expansion, accompanied by marked reduction in plasma membrane tension. These biophysical changes promoted the phagocytosis of bacteria, fungus, apoptotic corpses, and cancer cells. We also found that Gß4-deficient neutrophils are defective in the normal inhibition of migration following cargo uptake. Sphingolipid synthesis played a central role in these phenotypes by driving plasma membrane accumulation in cells lacking Gß4. In Gß4 knockout mice, neutrophils not only exhibited enhanced phagocytosis of inhaled fungal conidia in the lung but also increased trafficking of engulfed pathogens to other organs. Together, these results reveal an unexpected, biophysical control mechanism central to myeloid functional decision-making.


Subject(s)
Cell Membrane , Mice, Knockout , Phagocytosis , Animals , Phagocytosis/immunology , Cell Membrane/metabolism , Cell Membrane/immunology , Mice , Myeloid Cells/immunology , Mice, Inbred C57BL , Neutrophils/immunology , Macrophages/immunology
15.
Nat Commun ; 15(1): 5107, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877001

ABSTRACT

Inositol hexaphosphate (InsP6) is the major storage form of phosphorus in seeds. Reducing seed InsP6 content is a breeding objective in agriculture, as InsP6 negatively impacts animal nutrition and the environment. Nevertheless, how InsP6 accumulation is regulated remains largely unknown. Here, we identify a clade of receptor-like cytoplasmic kinases (RLCKs), named Inositol Polyphosphate-related Cytoplasmic Kinases 1-6 (IPCK1-IPCK6), deeply involved in InsP6 accumulation. The InsP6 concentration is dramatically reduced in seeds of ipck quadruple (T-4m/C-4m) and quintuple (C-5m) mutants, accompanied with the obviously increase of phosphate (Pi) concentration. The plasma membrane-localized IPCKs recruit IPK1 involved in InsP6 synthesis, and facilitate its binding and activity via phosphorylation of GRF 14-3-3 proteins. IPCKs also recruit IPK2s and PI-PLCs required for InsP4/InsP5 and InsP3 biosynthesis respectively, to form a potential IPCK-GRF-PLC-IPK2-IPK1 complex. Our findings therefore uncover a regulatory mechanism of InsP6 accumulation governed by IPCKs, shedding light on the mechanisms of InsP biosynthesis in eukaryotes.


Subject(s)
14-3-3 Proteins , Arabidopsis Proteins , Arabidopsis , Phytic Acid , 14-3-3 Proteins/metabolism , 14-3-3 Proteins/genetics , Phytic Acid/metabolism , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Mutation , Cell Membrane/metabolism , Gene Expression Regulation, Plant , Inositol Phosphates/metabolism
16.
Sci Rep ; 14(1): 13748, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38877068

ABSTRACT

Subcellular membranes have complex lipid and protein compositions, which give rise to organelle-specific membrane packing, fluidity, and permeability. Due to its exquisite solvent sensitivity, the lipophilic fluorescence dye Nile Red has been used extensively to study membrane packing and polarity. Further improvement of Nile Red can be achieved by introducing electron-donating or withdrawing functional groups. Here, we compare the potential of derivatives of Nile Red with such functional substitutions for super-resolution fluorescence microscopy of lipid packing in model membranes and living cells. All studied Nile Red derivatives exhibit cholesterol-dependent fluorescence changes in model membranes, as shown by spectrally resolved stimulated emission depletion (STED) microscopy. STED imaging of Nile Red probes in cells reveals lower membrane packing in fibroblasts from healthy subjects compared to those from patients suffering from Niemann Pick type C1 (NPC1) disease, a lysosomal storage disorder with accumulation of cholesterol and sphingolipids in late endosomes and lysosomes. We also find small but consistent changes in the fluorescence lifetime of the Nile Red derivatives in NPC1 cells, suggesting altered hydrogen-bonding capacity in their membranes. All Nile Red derivatives are essentially non-fluorescent in water but increase their brightness in membranes, allowing for their use in MINFLUX single molecule tracking experiments. Our study uncovers the potential of Nile Red probes with functional substitutions for nanoscopic membrane imaging.


Subject(s)
Fluorescent Dyes , Microscopy, Fluorescence , Oxazines , Oxazines/chemistry , Humans , Microscopy, Fluorescence/methods , Fluorescent Dyes/chemistry , Cholesterol/metabolism , Fibroblasts/metabolism , Cell Membrane/metabolism
17.
Sci Rep ; 14(1): 13754, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38877109

ABSTRACT

The twin-arginine translocation (Tat) system transports folded proteins across energized biological membranes in bacteria, plastids, and plant mitochondria. In Escherichia coli, the three membrane proteins TatA, TatB and TatC associate to enable Tat transport. While TatB and TatC together form complexes that bind Tat-dependently transported proteins, the TatA component is responsible for the permeabilization of the membrane during transport. With wild type Tat systems, the TatB- and TatC-containing Tat complexes TC1 and TC2 can be differentiated. Their TatA content has not been resolved, nor could they be assigned to any step of the translocation mechanism. It is therefore a key question of current Tat research to understand how TatA associates with Tat systems during transport. By analyzing affinity-purified Tat complexes with mutations in TatC that selectively enrich either TC1 or TC2, we now for the first time demonstrate that both Tat complexes associate with TatA, but the larger TC2 recruits significantly more TatA than the smaller TC1. Most TatA co-purified as multimeric clusters. Using site-specific photo cross-linking, we could detect TatA-TatC interactions only near TatC transmembrane helices 5 and 6. Substrate-binding did not change the interacting positions but affected the stability of the interaction, pointing to a substrate-induced conformational transition. Together, our findings indicate that TatA clusters associate with TatBC without being integrated into the complex by major rearrangements. The increased TatA affinity of the larger Tat complex TC2 suggests that functional assembly is advanced in this complex.


Subject(s)
Cell Membrane , Escherichia coli Proteins , Escherichia coli , Membrane Transport Proteins , Protein Transport , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Escherichia coli/genetics , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/chemistry , Cell Membrane/metabolism , Protein Folding , Protein Binding , Mutation
18.
Nat Commun ; 15(1): 5119, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879572

ABSTRACT

One open question in the biology of growth factor receptors is how a quantitative input (i.e., ligand concentration) is decoded by the cell to produce specific response(s). Here, we show that an EGFR endocytic mechanism, non-clathrin endocytosis (NCE), which is activated only at high ligand concentrations and targets receptor to degradation, requires a tripartite organelle platform involving the plasma membrane (PM), endoplasmic reticulum (ER) and mitochondria. At these contact sites, EGFR-dependent, ER-generated Ca2+ oscillations are sensed by mitochondria, leading to increased metabolism and ATP production. Locally released ATP is required for cortical actin remodeling and EGFR-NCE vesicle fission. The same biochemical circuitry is also needed for an effector function of EGFR, i.e., collective motility. The multiorganelle signaling platform herein described mediates direct communication between EGFR signaling and mitochondrial metabolism, and is predicted to have a broad impact on cell physiology as it is activated by another growth factor receptor, HGFR/MET.


Subject(s)
Adenosine Triphosphate , Endocytosis , Endoplasmic Reticulum , ErbB Receptors , Mitochondria , Signal Transduction , Mitochondria/metabolism , ErbB Receptors/metabolism , Endoplasmic Reticulum/metabolism , Humans , Adenosine Triphosphate/metabolism , Animals , Cell Membrane/metabolism , Calcium Signaling/physiology , Calcium/metabolism
19.
Molecules ; 29(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38893395

ABSTRACT

High concentrations of acrolein (2-propenal) are found in polluted air and cigarette smoke, and may also be generated endogenously. Acrolein is also associated with the induction and progression of many diseases. The high reactivity of acrolein towards the thiol and amino groups of amino acids may cause damage to cell proteins. Acrolein may be responsible for the induction of oxidative stress in cells. We hypothesized that acrolein may contribute to the protein damage in erythrocytes, leading to the disruption of the structure of cell membranes. The lipid membrane fluidity, membrane cytoskeleton, and osmotic fragility were measured for erythrocytes incubated with acrolein for 24 h. The levels of thiol, amino, and carbonyl groups were determined in cell membrane and cytosol proteins. The level of non-enzymatic antioxidant potential (NEAC) and TBARS was also measured. The obtained research results showed that the exposure of erythrocytes to acrolein causes changes in the cell membrane and cytosol proteins. Acrolein stiffens the cell membrane of erythrocytes and increases their osmotic sensitivity. Moreover, it has been shown that erythrocytes treated with acrolein significantly reduce the non-enzymatic antioxidant potential of the cytosol compared to the control.


Subject(s)
Acrolein , Cytosol , Erythrocyte Membrane , Erythrocytes , Acrolein/pharmacology , Acrolein/toxicity , Acrolein/metabolism , Cytosol/metabolism , Cytosol/drug effects , Erythrocytes/drug effects , Erythrocytes/metabolism , Humans , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Oxidative Stress/drug effects , Antioxidants/pharmacology , Antioxidants/metabolism , Membrane Proteins/metabolism , Cell Membrane/metabolism , Cell Membrane/drug effects , Membrane Fluidity/drug effects , Osmotic Fragility/drug effects
20.
Cell Mol Life Sci ; 81(1): 272, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900158

ABSTRACT

We addressed the heteromerization of the epidermal growth factor receptor (EGFR) with G-protein coupled receptors (GPCR) on the basis of angiotensin-II-receptor-subtype-1(AT1R)-EGFR interaction as proof-of-concept and show its functional relevance during synergistic nuclear information transfer, beyond ligand-dependent EGFR transactivation. Following in silico modelling, we generated EGFR-interaction deficient AT1R-mutants and compared them to AT1R-wildtype. Receptor interaction was assessed by co-immunoprecipitation (CoIP), Förster resonance energy transfer (FRET) and fluorescence-lifetime imaging microscopy (FLIM). Changes in cell morphology, ERK1/2-phosphorylation (ppERK1/2), serum response factor (SRF)-activation and cFOS protein expression were determined by digital high content microscopy at the single cell level. FRET, FLIM and CoIP confirmed the physical interaction of AT1R-wildtype with EGFR that was strongly reduced for the AT1R-mutants. Responsiveness of cells transfected with AT1R-WT or -mutants to angiotensin II or EGF was similar regarding changes in cell circularity, ppERK1/2 (direct and by ligand-dependent EGFR-transactivation), cFOS-expression and SRF-activity. By contrast, the EGFR-AT1R-synergism regarding these parameters was completely absent for in the interaction-deficient AT1R mutants. The results show that AT1R-EGFR heteromerisation enables AT1R-EGFR-synergism on downstream gene expression regulation, modulating the intensity and the temporal pattern of nuclear AT1R/EGFR-information transfer. Furthermore, remote EGFR transactivation, via ligand release or cytosolic tyrosine kinases, is not sufficient for the complete synergistic control of gene expression.


Subject(s)
Cell Nucleus , ErbB Receptors , Receptor, Angiotensin, Type 1 , ErbB Receptors/metabolism , Humans , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/genetics , Cell Nucleus/metabolism , Fluorescence Resonance Energy Transfer , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Cell Membrane/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Epidermal Growth Factor/metabolism , HEK293 Cells , Protein Binding , Serum Response Factor/metabolism , Serum Response Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL