Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 404
Filter
1.
ACS Appl Mater Interfaces ; 15(43): 49988-50001, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37862704

ABSTRACT

Cell surface receptors facilitate signaling and nutrient uptake. These processes are dynamic, requiring receptors to be actively recycled by endocytosis. Due to their differential expression in disease states, receptors are often the target of drug-carrier particles, which are adorned with ligands that bind specifically to receptors. These targeted particles are taken into the cell by multiple routes of internalization, where the best-characterized pathway is clathrin-mediated endocytosis. Most studies of particle uptake have utilized bulk assays rather than observing individual endocytic events. As a result, the detailed mechanisms of particle uptake remain obscure. To address this gap, we employed a live-cell imaging approach to study the uptake of individual liposomes as they interact with clathrin-coated structures. By tracking individual internalization events, we find that the size of liposomes rather than the density of the ligands on their surfaces primarily determines their probability of uptake. Interestingly, targeting has the greatest impact on endocytosis of liposomes of intermediate diameters, with the smallest and largest liposomes being internalized or excluded, respectively, regardless of whether they are targeted. These findings, which highlight a previously unexplored limitation of targeted delivery, can be used to design more effective drug carriers.


Subject(s)
Endocytosis , Liposomes , Liposomes/chemistry , Drug Carriers/pharmacology , Biological Transport , Clathrin/chemistry
2.
J Biol Chem ; 299(9): 105091, 2023 09.
Article in English | MEDLINE | ID: mdl-37516240

ABSTRACT

α-Synuclein and family members ß- and γ-synuclein are presynaptic proteins that sense and generate membrane curvature, properties important for synaptic vesicle (SV) cycling. αßγ-synuclein triple knockout neurons exhibit SV endocytosis deficits. Here, we investigated if α-synuclein affects clathrin assembly in vitro. Visualizing clathrin assembly on membranes using a lipid monolayer system revealed that α-synuclein increases clathrin lattices size and curvature. On cell membranes, we observe that α-synuclein is colocalized with clathrin and its adapter AP180 in a concentric ring pattern. Clathrin puncta that contain both α-synuclein and AP180 were significantly larger than clathrin puncta containing either protein alone. We determined that this effect occurs in part through colocalization of α-synuclein with the phospholipid PI(4,5)P2 in the membrane. Immuno-electron microscopy (EM) of synaptosomes uncovered that α-synuclein relocalizes from SVs to the presynaptic membrane upon stimulation, positioning α-synuclein to function on presynaptic membranes during or after stimulation. Additionally, we show that deletion of synucleins impacts brain-derived clathrin-coated vesicle size. Thus, α-synuclein affects the size and curvature of clathrin structures on membranes and functions as an endocytic accessory protein.


Subject(s)
Clathrin , Monomeric Clathrin Assembly Proteins , alpha-Synuclein , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Cell Membrane/metabolism , Clathrin/chemistry , Clathrin/metabolism , Endocytosis , Microscopy, Immunoelectron , Monomeric Clathrin Assembly Proteins/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Synaptosomes/metabolism , Protein Transport , In Vitro Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Brain/cytology , Clathrin-Coated Vesicles/metabolism
3.
Curr Opin Struct Biol ; 75: 102427, 2022 08.
Article in English | MEDLINE | ID: mdl-35872561

ABSTRACT

Clathrin-mediated endocytosis enables selective uptake of molecules into cells in response to changing cellular needs. It occurs through assembly of coat components around the plasma membrane that determine vesicle contents and facilitate membrane bending to form a clathrin-coated transport vesicle. In this review we discuss recent cryo-electron microscopy structures that have captured a series of events in the life cycle of a clathrin-coated vesicle. Both single particle analysis and tomography approaches have revealed details of the clathrin lattice structure itself, how AP2 may interface with clathrin within a coated vesicle and the importance of PIP2 binding for assembly of the yeast adaptors Sla2 and Ent1 on the membrane. Within cells, cryo-electron tomography of clathrin in flat lattices and high-speed AFM studies provided new insights into how clathrin morphology can adapt during CCV formation. Thus, key mechanical processes driving clathrin-mediated endocytosis have been captured through multiple techniques working in partnership.


Subject(s)
Clathrin , Endocytosis , Cell Membrane/metabolism , Clathrin/chemistry , Clathrin/metabolism , Clathrin-Coated Vesicles/metabolism , Coated Vesicles/metabolism , Cryoelectron Microscopy , Saccharomyces cerevisiae/metabolism
4.
Nanoscale ; 14(28): 10200-10210, 2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35796347

ABSTRACT

To detect heavy metal toxicity using self-assembled nanostructures, a clathrin triskelion-inspired highly functional C3-symmetric trimerized biotinylated di-tryptophan peptide was used. This triskelion peptide is known to self-assemble into nanotorus-like structures and can therefore act as a nanocage for various analytes. In this work, in addition to spectroscopy, force and electron microscopy were successfully used to detect the effect of toxic metal ions such as zinc, cadmium, and mercury by exploiting the change in the nanotorus morphology. Different concentrations of mercury led to the expansion of nanotorus structures into microtori. Therefore, we provide a unique application of heavy metal toxicity by utilizing "material nanoarchitectonics" to architect nanotorus structures into higher-order microtorus structures, as instructed by mercury. Such a strategy can make heavy metal sensing easier for materials scientists and open new avenues for biomedical/environmental science applications.


Subject(s)
Mercury , Nanostructures , Cadmium , Clathrin/chemistry , Peptides
5.
PLoS Comput Biol ; 18(3): e1009969, 2022 03.
Article in English | MEDLINE | ID: mdl-35312692

ABSTRACT

Clathrin-coated structures must assemble on cell membranes to internalize receptors, with the clathrin protein only linked to the membrane via adaptor proteins. These structures can grow surprisingly large, containing over 20 clathrin, yet they often fail to form productive vesicles, instead aborting and disassembling. We show that clathrin structures of this size can both form and disassemble spontaneously when adaptor protein availability is low, despite high abundance of clathrin. Here, we combine recent in vitro kinetic measurements with microscopic reaction-diffusion simulations and theory to differentiate mechanisms of stable vs unstable clathrin assembly on membranes. While in vitro conditions drive assembly of robust, stable lattices, we show that concentrations, geometry, and dimensional reduction in physiologic-like conditions do not support nucleation if only the key adaptor AP-2 is included, due to its insufficient abundance. Nucleation requires a stoichiometry of adaptor to clathrin that exceeds 1:1, meaning additional adaptor types are necessary to form lattices successfully and efficiently. We show that the critical nucleus contains ~25 clathrin, remarkably similar to sizes of the transient and abortive structures observed in vivo. Lastly, we quantify the cost of bending the membrane under our curved clathrin lattices using a continuum membrane model. We find that the cost of bending the membrane could be largely offset by the energetic benefit of forming curved rather than flat structures, with numbers comparable to experiments. Our model predicts how adaptor density can tune clathrin-coated structures from the transient to the stable, showing that active energy consumption is therefore not required for lattice disassembly or remodeling during growth, which is a critical advance towards predicting productive vesicle formation.


Subject(s)
Clathrin , Endocytosis , Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/metabolism , Clathrin/chemistry
6.
Nat Commun ; 13(1): 905, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35173166

ABSTRACT

The crosstalk between growth factor and adhesion receptors is key for cell growth and migration. In pathological settings, these receptors are drivers of cancer. Yet, how growth and adhesion signals are spatially organized and integrated is poorly understood. Here we use quantitative fluorescence and electron microscopy to reveal a mechanism where flat clathrin lattices partition and activate growth factor signals via a coordinated response that involves crosstalk between epidermal growth factor receptor (EGFR) and the adhesion receptor ß5-integrin. We show that ligand-activated EGFR, Grb2, Src, and ß5-integrin are captured by clathrin coated-structures at the plasma membrane. Clathrin structures dramatically grow in response to EGF into large flat plaques and provide a signaling platform that link EGFR and ß5-integrin through Src-mediated phosphorylation. Disrupting this EGFR/Src/ß5-integrin axis prevents both clathrin plaque growth and dampens receptor signaling. Our study reveals a reciprocal regulation between clathrin lattices and two different receptor systems to coordinate and enhance signaling. These findings have broad implications for the regulation of growth factor signaling, adhesion, and endocytosis.


Subject(s)
Clathrin-Coated Vesicles/metabolism , Clathrin/chemistry , GRB2 Adaptor Protein/metabolism , Integrin beta Chains/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Endocytosis , ErbB Receptors/metabolism , Humans , Microscopy, Electron , Signal Transduction/physiology , src-Family Kinases/metabolism
7.
Biochem Biophys Res Commun ; 587: 69-77, 2022 01 08.
Article in English | MEDLINE | ID: mdl-34864549

ABSTRACT

The clathrin coat assembly protein AP180 drives endocytosis, which is crucial for numerous physiological events, such as the internalization and recycling of receptors, uptake of neurotransmitters and entry of viruses, including SARS-CoV-2, by interacting with clathrin. Moreover, dysfunction of AP180 underlies the pathogenesis of Alzheimer's disease. Therefore, it is important to understand the mechanisms of assembly and, especially, disassembly of AP180/clathrin-containing cages. Here, we identified AP180 as a novel phosphatidic acid (PA)-binding protein from the mouse brain. Intriguingly, liposome binding assays using various phospholipids and PA species revealed that AP180 most strongly bound to 1-stearoyl-2-docosahexaenoyl-PA (18:0/22:6-PA) to a comparable extent as phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), which is known to associate with AP180. An AP180 N-terminal homology domain (1-289 aa) interacted with 18:0/22:6-PA, and a lysine-rich motif (K38-K39-K40) was essential for binding. The 18:0/22:6-PA in liposomes in 100 nm diameter showed strong AP180-binding activity at neutral pH. Notably, 18:0/22:6-PA significantly attenuated the interaction of AP180 with clathrin. However, PI(4,5)P2 did not show such an effect. Taken together, these results indicate the novel mechanism by which 18:0/22:6-PA selectively regulates the disassembly of AP180/clathrin-containing cages.


Subject(s)
Clathrin/metabolism , Docosahexaenoic Acids/metabolism , Monomeric Clathrin Assembly Proteins/metabolism , Phosphatidic Acids/metabolism , Animals , Binding Sites , Brain/metabolism , COVID-19/metabolism , COVID-19/virology , Cell Line , Clathrin/chemistry , Docosahexaenoic Acids/chemistry , Endocytosis/physiology , Host Microbial Interactions/physiology , Humans , Mice , Monomeric Clathrin Assembly Proteins/chemistry , Monomeric Clathrin Assembly Proteins/genetics , Phosphatidic Acids/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SARS-CoV-2/physiology , Virus Internalization
9.
Neuron ; 109(18): 2884-2901.e7, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34534453

ABSTRACT

In non-neuronal cells, clathrin has established roles in endocytosis, with clathrin cages enclosing plasma membrane infoldings, followed by rapid disassembly and reuse of monomers. However, in neurons, clathrin is conveyed in slow axonal transport over days to weeks, and the underlying transport/targeting mechanisms, mobile cargo structures, and even its precise presynaptic localization and physiologic role are unclear. Combining live imaging, photobleaching/conversion, mass spectrometry, electron microscopy, and super-resolution imaging, we found that unlike in dendrites, where clathrin cages rapidly assemble and disassemble, in axons, clathrin and related proteins organize into stable "transport packets" that are unrelated to endocytosis and move intermittently on microtubules, generating an overall slow anterograde flow. At synapses, multiple clathrin packets abut synaptic vesicle (SV) clusters, and clathrin packets also exchange between synaptic boutons in a microtubule-dependent "superpool." Within synaptic boundaries, clathrin is surprisingly dynamic, continuously exchanging between local clathrin assemblies, and its depletion impairs SV recycling. Our data provide a conceptual framework for understanding clathrin trafficking and presynaptic targeting that has functional implications.


Subject(s)
Axonal Transport/physiology , Clathrin-Coated Vesicles/metabolism , Clathrin/metabolism , Hippocampus/metabolism , Synapses/metabolism , Animals , Animals, Newborn , Cells, Cultured , Clathrin/chemistry , Clathrin-Coated Vesicles/chemistry , Hippocampus/chemistry , Hippocampus/cytology , Mice , Protein Transport/physiology , Rats , Rats, Wistar , Synapses/chemistry , Time-Lapse Imaging/methods
10.
Biochim Biophys Acta Biomembr ; 1863(12): 183757, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34478732

ABSTRACT

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


Subject(s)
Carrier Proteins/genetics , Glucose Transporter Type 1/genetics , Lipids/genetics , Phosphatidylinositol 4,5-Diphosphate/chemistry , Biological Transport/genetics , Carrier Proteins/chemistry , Clathrin/chemistry , Endocytosis/genetics , Glucose/metabolism , Glucose Transporter Type 1/chemistry , Humans , Lipids/chemistry , Phosphatidylinositol 4,5-Diphosphate/genetics , Signal Transduction
11.
EMBO J ; 40(19): e108795, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34487371

ABSTRACT

Clathrin-coated pits are formed by the recognition of membrane and cargo by the AP2 complex and the subsequent recruitment of clathrin triskelia. A role for AP2 in coated-pit assembly beyond initial clathrin recruitment has not been explored. Clathrin binds the ß2 subunit of AP2, and several binding sites have been identified, but our structural knowledge of these interactions is incomplete and their functional importance during endocytosis is unclear. Here, we analysed the cryo-EM structure of clathrin cages assembled in the presence of ß2 hinge-appendage (ß2HA). We find that the ß2-appendage binds in at least two positions in the cage, demonstrating that multi-modal binding is a fundamental property of clathrin-AP2 interactions. In one position, ß2-appendage cross-links two adjacent terminal domains from different triskelia. Functional analysis of ß2HA-clathrin interactions reveals that endocytosis requires two clathrin interaction sites: a clathrin-box motif on the hinge and the "sandwich site" on the appendage. We propose that ß2-appendage binding to more than one triskelion is a key feature of the system and likely explains why assembly is driven by AP2.


Subject(s)
Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Clathrin/chemistry , Clathrin/metabolism , Coated Vesicles/chemistry , Coated Vesicles/metabolism , Models, Molecular , Amino Acid Sequence , Binding Sites , Coated Pits, Cell-Membrane/chemistry , Coated Pits, Cell-Membrane/metabolism , Endocytosis , Fluorescent Antibody Technique , HeLa Cells , Humans , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Structure-Activity Relationship
12.
Biochem J ; 478(16): 3099-3123, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34436540

ABSTRACT

Clathrin is best known for its contribution to clathrin-mediated endocytosis yet it also participates to a diverse range of cellular functions. Key to this is clathrin's ability to assemble into polyhedral lattices that include curved football or basket shapes, flat lattices or even tubular structures. In this review, we discuss clathrin structure and coated vesicle formation, how clathrin is utilised within different cellular processes including synaptic vesicle recycling, hormone desensitisation, spermiogenesis, cell migration and mitosis, and how clathrin's remarkable 'shapeshifting' ability to form diverse lattice structures might contribute to its multiple cellular functions.


Subject(s)
Cell Membrane/metabolism , Clathrin/metabolism , Endocytosis , Endosomes/metabolism , Exocytosis , Animals , Clathrin/chemistry , Clathrin/ultrastructure , Humans , Microscopy, Electron/methods , Models, Biological , Protein Conformation
13.
PLoS Comput Biol ; 17(1): e1008474, 2021 01.
Article in English | MEDLINE | ID: mdl-33406091

ABSTRACT

Intrinsically disordered regions (IDRs) are prevalent in the eukaryotic proteome. Common functional roles of IDRs include forming flexible linkers or undergoing allosteric folding-upon-binding. Recent studies have suggested an additional functional role for IDRs: generating steric pressure on the plasma membrane during endocytosis, via molecular crowding. However, in order to accomplish useful functions, such crowding needs to be regulated in space (e.g., endocytic hotspots) and time (e.g., during vesicle formation). In this work, we explore binding-induced regulation of IDR steric volume. We simulate the IDRs of two proteins from Clathrin-mediated endocytosis (CME) to see if their conformational spaces are regulated via binding-induced expansion. Using Monte-Carlo computational modeling of excluded volumes, we generate large conformational ensembles (3 million) for the IDRs of Epsin and Eps15 and dock the conformers to the alpha subunit of Adaptor Protein 2 (AP2α), their CME binding partner. Our results show that as more molecules of AP2α are bound, the Epsin-derived ensemble shows a significant increase in global dimensions, measured as the radius of Gyration (RG) and the end-to-end distance (EED). Unlike Epsin, Eps15-derived conformers that permit AP2α binding at one motif were found to be more likely to accommodate binding of AP2α at other motifs, suggesting a tendency toward co-accessibility of binding motifs. Co-accessibility was not observed for any pair of binding motifs in Epsin. Thus, we speculate that the disordered regions of Epsin and Eps15 perform different roles during CME, with accessibility in Eps15 allowing it to act as a recruiter of AP2α molecules, while binding-induced expansion of the Epsin disordered region could impose steric pressure and remodel the plasma membrane during vesicle formation.


Subject(s)
Adaptor Protein Complex 2 , Adaptor Proteins, Vesicular Transport , Intrinsically Disordered Proteins , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex 2/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Clathrin/chemistry , Clathrin/metabolism , Endocytosis/physiology , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Molecular Docking Simulation , Protein Binding , Protein Conformation
14.
Methods Mol Biol ; 2233: 71-91, 2021.
Article in English | MEDLINE | ID: mdl-33222128

ABSTRACT

Endocytosis is the dynamic internalization of cargo (receptors, hormones, viruses) for cellular signaling or processing. It involves multiple mechanisms, classified depending on critical proteins involved, speed, morphology of the derived intracellular vesicles, or substance trafficked. Pharmacological targeting of specific endocytosis pathways has a proven utility for diverse clinical applications from epilepsy to cancer. A multiplexable, high-content screening assay has been designed and implemented to assess various forms of endocytic trafficking and the associated impact of potential small molecule modulators. The applications of this assay include (1) drug discovery in the search for specific, cell-permeable endocytosis pathway inhibitors (and associated analogues from structure-activity relationship studies), (2) deciphering the mechanism of internalization for a novel ligand (using pathway-specific inhibitors), (3) assessment of the importance of specific proteins in the trafficking process (using CRISPR-Cas9 technology, siRNA treatment, or transfection), and (4) identifying whether endocytosis inhibition is an off-target for novel compounds designed for alternative purposes. We describe this method in detail and provide a range of troubleshooting options and alternatives to modify the protocol for lab-specific applications.


Subject(s)
Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Endocytosis/drug effects , Signal Transduction/drug effects , Cell Line, Tumor , Clathrin/chemistry , Humans , Ligands
15.
Nat Methods ; 17(11): 1097-1099, 2020 11.
Article in English | MEDLINE | ID: mdl-33046895

ABSTRACT

vLUME is a virtual reality software package designed to render large three-dimensional single-molecule localization microscopy datasets. vLUME features include visualization, segmentation, bespoke analysis of complex local geometries and exporting features. vLUME can perform complex analysis on real three-dimensional biological samples that would otherwise be impossible by using regular flat-screen visualization programs.


Subject(s)
Image Processing, Computer-Assisted/methods , Single Molecule Imaging/methods , Virtual Reality , Algorithms , Animals , COS Cells , Caulobacter crescentus/chemistry , Cell Line , Cell Membrane/chemistry , Chlorocebus aethiops , Clathrin/chemistry , Humans , Jurkat Cells , Microtubules/chemistry , Nuclear Pore/chemistry , Software
16.
Biochim Biophys Acta Mol Cell Res ; 1867(12): 118849, 2020 12.
Article in English | MEDLINE | ID: mdl-32916203

ABSTRACT

FPR2, a member of the family of G protein-coupled receptors (GPCRs), mediates neutrophil migration, a response that has been linked to ß-arrestin recruitment. ß-Arrestin regulates GPCR endocytosis and can also elicit non-canonical receptor signaling. To determine the poorly understood role of ß-arrestin in FPR2 endocytosis and in NADPH-oxidase activation in neutrophils, Barbadin was used as a research tool in this study. Barbadin has been shown to bind the clathrin adaptor protein (AP2) and thereby prevent ß-arrestin/AP2 interaction and ß-arrestin-mediated GPCR endocytosis. In agreement with this, AP2/ß-arrestin interaction induced by an FPR2-specific agonist was inhibited by Barbadin. Unexpectedly, however, Barbadin did not inhibit FPR2 endocytosis, indicating that a mechanism independent of ß-arrestin/AP2 interaction may sustain FPR2 endocytosis. This was confirmed by the fact, that FPR2 also underwent agonist-promoted endocytosis in ß-arrestin deficient cells, albeit at a diminished level as compared to wild type cells. Dissection of the Barbadin effects on FPR2-mediated neutrophil functions including NADPH-oxidase activation mediated release of reactive oxygen species (ROS) and chemotaxis revealed that Barbadin had no effect on chemotactic migration whereas the release of ROS was potentiated/primed. The effect of Barbadin on ROS production was reversible, independent of ß-arrestin recruitment, and similar to that induced by latrunculin A. Taken together, our data demonstrate that endocytic uptake of FPR2 occurs independently of ß-arrestin, while Barbadin selectively augments FPR2-mediated ROS production independently of receptor endocytosis. Given that Barbadin binds to AP2 and prevents the AP2/ß-arrestin interaction, our results indicate a role for AP2 in FPR2-mediated ROS release from neutrophils.


Subject(s)
Endocytosis/genetics , Pyrimidines/pharmacology , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , beta-Arrestin 1/genetics , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex 2/genetics , Clathrin/chemistry , Endocytosis/drug effects , HEK293 Cells , Humans , NADPH Oxidases/genetics , Neutrophils/drug effects , Protein Binding/drug effects , Pyrimidines/chemistry , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Lipoxin/chemistry , Signal Transduction/drug effects , beta-Arrestin 1/chemistry
17.
J Microencapsul ; 37(8): 566-576, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32928025

ABSTRACT

AIM: MUC-1-peptide (M-1-pep) loaded poly (lactide-co-glycolide) nanoparticles were coated with protamine sulphate (PS), M-1-pep-PS-P-NPs for targeting antigen presenting cells (APCs) to evoke cytokine release. METHODS AND RESULTS: M-1-pep-PS-P-NPs were tailored by emulsion-diffusion evaporation method and characterised in vitro under a set of rigorous parameters. The average particle size and zeta potential of optimised M-1-pep-PS-P-B-NPs was measured to be 132.21 ± 30.71 nm and 6.29 ± 0.71 mV, significantly (p < 0.01) higher than 71.24 ± 17.76-nm and -43.41 ± 3.37 mV of M-1-pep-P-NPs. Further, 50-µg/ml concentration of M-1-pep-PS-P-B-NPs displayed 82.4% cellular uptake in RAW 264.7 cells calculated in setting of fluorescence intensity significantly (p < 0.05) elevated than 63.1% of M-1-pep-P-NPs. Consistent to quantitative results, M-1-pep-PS-P-B-NPs also confirmed advanced cellular uptake (CU) in RAW 264.7 cells in contrast to M-1-pep-P-NPs suppose to be through multiple mechanisms including phagocytosis and clathrin mediated endocytosis. CONCLUSION: M-1-pep-PS-P-B-NPs must be evaluated in vivo through inhalation route of administration for antitumor prospective in lung cancer xenograft model.


Subject(s)
Cytokines/metabolism , Mucin-1/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Antigens/chemistry , Clathrin/chemistry , Diffusion , Endocytosis , In Vitro Techniques , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Neoplasm Transplantation , Particle Size , Phagocytosis , RAW 264.7 Cells , Signal Transduction
18.
Curr Opin Cell Biol ; 65: 141-149, 2020 08.
Article in English | MEDLINE | ID: mdl-32836101

ABSTRACT

Understanding of the range and mechanisms of clathrin functions has developed exponentially since clathrin's discovery in 1975. Here, newly established molecular mechanisms that regulate clathrin activity and connect clathrin pathways to differentiation, disease and physiological processes such as glucose metabolism are reviewed. Diversity and commonalities of clathrin pathways across the tree of life reveal species-specific differences enabling functional plasticity in both membrane traffic and cytokinesis. New structural information on clathrin coat formation and cargo interactions emphasises the interplay between clathrin, adaptor proteins, lipids and cargo, and how this interplay regulates quality control of clathrin's function and is compromised in infection and neurological disease. Roles for balancing clathrin-mediated cargo transport are defined in stem cell development and additional disease states.


Subject(s)
Clathrin/metabolism , Disease , Animals , Biological Transport , Clathrin/chemistry , Humans , Models, Biological , Organ Specificity , Phylogeny
19.
Sci Rep ; 10(1): 12773, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32728195

ABSTRACT

RNA interference (RNAi) technologies have recently been developed to control a growing number of agronomically significant fungal phytopathogens, including the white mold pathogen, Sclerotinia sclerotiorum. Exposure of this fungus to exogenous double-stranded RNA (dsRNA) results in potent RNAi-mediated knockdown of target genes' transcripts, but it is unclear how the dsRNA can enter the fungal cells. In nematodes, specialized dsRNA transport proteins such as SID-1 facilitate dsRNA uptake, but for many other eukaryotes in which the dsRNA uptake mechanisms have been examined, endocytosis appears to mediate the uptake process. In this study, using live cell imaging, transgenic fungal cultures and endocytic inhibitors, we determined that the uptake mechanism in S. sclerotiorum occurs through clathrin-mediated endocytosis. RNAi-mediated knockdown of several clathrin-mediated endocytic genes' transcripts confirmed the involvement of this cellular uptake process in facilitating RNAi in this fungus. Understanding the mode of dsRNA entry into the fungus will prove useful in designing and optimizing future dsRNA-based control methods and in anticipating possible mechanisms by which phytopathogens may develop resistance to this novel category of fungicides.


Subject(s)
Ascomycota/metabolism , Clathrin/chemistry , Endocytosis , RNA Interference , RNA, Double-Stranded/chemistry , Animals , Biological Transport , CHO Cells , Cricetulus , Cytoplasm/metabolism , Green Fluorescent Proteins/metabolism , Microscopy, Confocal , Transgenes
20.
J Mol Cell Cardiol ; 144: 127-139, 2020 07.
Article in English | MEDLINE | ID: mdl-32445844

ABSTRACT

Ion channel trafficking powerfully influences cardiac electrical activity as it regulates the number of available channels at the plasma membrane. Studies have largely focused on identifying the molecular determinants of the trafficking of the atria-specific KV1.5 channel, the molecular basis of the ultra-rapid delayed rectifier current IKur. Besides, regulated KV1.5 channel recycling upon changes in homeostatic state and mechanical constraints in native cardiomyocytes has been well documented. Here, using cutting-edge imaging in live myocytes, we investigated the dynamics of this channel in the plasma membrane. We demonstrate that the clathrin pathway is a major regulator of the functional expression of KV1.5 channels in atrial myocytes, with the microtubule network as the prominent organizer of KV1.5 transport within the membrane. Both clathrin blockade and microtubule disruption result in channel clusterization with reduced membrane mobility and internalization, whereas disassembly of the actin cytoskeleton does not. Mobile KV1.5 channels are associated with the microtubule plus-end tracking protein EB1 whereas static KV1.5 clusters are associated with stable acetylated microtubules. In human biopsies from patients in atrial fibrillation associated with atrial remodeling, drastic modifications in the trafficking balance occurs together with alteration in microtubule polymerization state resulting in modest reduced endocytosis and increased recycling. Consequently, hallmark of atrial KV1.5 dynamics within the membrane is clathrin- and microtubule- dependent. During atrial remodeling, predominance of anterograde trafficking activity over retrograde trafficking could result in accumulation ok KV1.5 channels in the plasma membrane.


Subject(s)
Clathrin/metabolism , Microtubules/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Multimerization , Animals , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Atrial Remodeling/genetics , Clathrin/chemistry , Clathrin-Coated Vesicles , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Electrophysiological Phenomena , Heart Atria/metabolism , Humans , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Microtubules/chemistry , Microtubules/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Potassium Channels, Voltage-Gated/chemistry , Rats , Sarcolemma/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...