Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.802
Filter
1.
Annu Rev Immunol ; 36: 103-125, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29261409

ABSTRACT

T cell receptors (TCRs) are protein complexes formed by six different polypeptides. In most T cells, TCRs are composed of αß subunits displaying immunoglobulin-like variable domains that recognize peptide antigens associated with major histocompatibility complex molecules expressed on the surface of antigen-presenting cells. TCRαß subunits are associated with the CD3 complex formed by the γ, δ, ε, and ζ subunits, which are invariable and ensure signal transduction. Here, we review how the expression and function of TCR complexes are orchestrated by several fine-tuned cellular processes that encompass (a) synthesis of the subunits and their correct assembly and expression at the plasma membrane as a single functional complex, (b) TCR membrane localization and dynamics at the plasma membrane and in endosomal compartments, (c) TCR signal transduction leading to T cell activation, and (d) TCR degradation. These processes balance each other to ensure efficient T cell responses to a variety of antigenic stimuli while preventing autoimmunity.


Subject(s)
Gene Expression Regulation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Biomarkers , CD3 Complex/genetics , CD3 Complex/metabolism , Cell Membrane/metabolism , Endocytosis/genetics , Endocytosis/immunology , Endosomes/metabolism , Humans , Immunomodulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Proteolysis , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Structure-Activity Relationship
2.
Annu Rev Immunol ; 36: 717-753, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29490164

ABSTRACT

Antigen cross-presentation is an adaptation of the cellular process of loading MHC-I molecules with endogenous peptides during their biosynthesis within the endoplasmic reticulum. Cross-presented peptides derive from internalized proteins, microbial pathogens, and transformed or dying cells. The physical separation of internalized cargo from the endoplasmic reticulum, where the machinery for assembling peptide-MHC-I complexes resides, poses a challenge. To solve this problem, deliberate rewiring of organelle communication within cells is necessary to prepare for cross-presentation, and different endocytic receptors and vesicular traffic patterns customize the emergent cross-presentation compartment to the nature of the peptide source. Three distinct pathways of vesicular traffic converge to form the ideal cross-presentation compartment, each regulated differently to supply a unique component that enables cross-presentation of a diverse repertoire of peptides. Delivery of centerpiece MHC-I molecules is the critical step regulated by microbe-sensitive Toll-like receptors. Defining the subcellular sources of MHC-I and identifying sites of peptide loading during cross-presentation remain key challenges.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Cross-Priming/immunology , Immunomodulation , Animals , Biological Transport , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endocytosis/immunology , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Epitopes/immunology , Epitopes/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Intracellular Space/metabolism , Phagocytosis/immunology , Proteolysis , Receptors, Cell Surface/metabolism
3.
Cell ; 187(2): 312-330.e22, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38157854

ABSTRACT

The FERONIA (FER)-LLG1 co-receptor and its peptide ligand RALF regulate myriad processes for plant growth and survival. Focusing on signal-induced cell surface responses, we discovered that intrinsically disordered RALF triggers clustering and endocytosis of its cognate receptors and FER- and LLG1-dependent endocytosis of non-cognate regulators of diverse processes, thus capable of broadly impacting downstream responses. RALF, however, remains extracellular. We demonstrate that RALF binds the cell wall polysaccharide pectin. They phase separate and recruit FER and LLG1 into pectin-RALF-FER-LLG1 condensates to initiate RALF-triggered cell surface responses. We show further that two frequently encountered environmental challenges, elevated salt and temperature, trigger RALF-pectin phase separation, promiscuous receptor clustering and massive endocytosis, and that this process is crucial for recovery from stress-induced growth attenuation. Our results support that RALF-pectin phase separation mediates an exoskeletal mechanism to broadly activate FER-LLG1-dependent cell surface responses to mediate the global role of FER in plant growth and survival.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Phosphotransferases/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Pectins/metabolism , Phase Separation , GPI-Linked Proteins/metabolism
4.
Cell ; 186(4): 821-836.e13, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36750096

ABSTRACT

The low-density lipoprotein (LDL) receptor-related protein 2 (LRP2 or megalin) is representative of the phylogenetically conserved subfamily of giant LDL receptor-related proteins, which function in endocytosis and are implicated in diseases of the kidney and brain. Here, we report high-resolution cryoelectron microscopy structures of LRP2 isolated from mouse kidney, at extracellular and endosomal pH. The structures reveal LRP2 to be a molecular machine that adopts a conformation for ligand binding at the cell surface and for ligand shedding in the endosome. LRP2 forms a homodimer, the conformational transformation of which is governed by pH-sensitive sites at both homodimer and intra-protomer interfaces. A subset of LRP2 deleterious missense variants in humans appears to impair homodimer assembly. These observations lay the foundation for further understanding the function and mechanism of LDL receptors and implicate homodimerization as a conserved feature of the LRP receptor subfamily.


Subject(s)
Endocytosis , Low Density Lipoprotein Receptor-Related Protein-2 , Animals , Humans , Mice , Cryoelectron Microscopy , Kidney/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism
5.
Cell ; 185(24): 4560-4573.e19, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36368322

ABSTRACT

Binding of arrestin to phosphorylated G protein-coupled receptors (GPCRs) is crucial for modulating signaling. Once internalized, some GPCRs remain complexed with ß-arrestins, while others interact only transiently; this difference affects GPCR signaling and recycling. Cell-based and in vitro biophysical assays reveal the role of membrane phosphoinositides (PIPs) in ß-arrestin recruitment and GPCR-ß-arrestin complex dynamics. We find that GPCRs broadly stratify into two groups, one that requires PIP binding for ß-arrestin recruitment and one that does not. Plasma membrane PIPs potentiate an active conformation of ß-arrestin and stabilize GPCR-ß-arrestin complexes by promoting a fully engaged state of the complex. As allosteric modulators of GPCR-ß-arrestin complex dynamics, membrane PIPs allow for additional conformational diversity beyond that imposed by GPCR phosphorylation alone. For GPCRs that require membrane PIP binding for ß-arrestin recruitment, this provides a mechanism for ß-arrestin release upon translocation of the GPCR to endosomes, allowing for its rapid recycling.


Subject(s)
Arrestins , Phosphatidylinositols , beta-Arrestins/metabolism , Phosphatidylinositols/metabolism , Arrestins/metabolism , beta-Arrestin 1/metabolism , Receptors, G-Protein-Coupled/metabolism
6.
Annu Rev Biochem ; 90: 709-737, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33606955

ABSTRACT

Intricate relationships between endocytosis and cellular signaling, first recognized nearly 40 years ago through the study of tyrosine kinase growth factor receptors, are now known to exist for multiple receptor classes and to affect myriad physiological and developmental processes. This review summarizes our present understanding of how endocytosis orchestrates cellular signaling networks, with an emphasis on mechanistic underpinnings and focusing on two receptor classes-tyrosine kinase and G protein-coupled receptors-that have been investigated in particular detail. Together, these examples provide a useful survey of the current consensus, uncertainties, and controversies in this rapidly advancing area of cell biology.


Subject(s)
Endocytosis/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Membrane/metabolism , Endosomes/metabolism , Humans , Lysosomes/metabolism , Protein Transport , Signal Transduction
7.
Cell ; 180(5): 895-914.e27, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32142680

ABSTRACT

A safe and controlled manipulation of endocytosis in vivo may have disruptive therapeutic potential. Here, we demonstrate that the anti-emetic/anti-psychotic prochlorperazine can be repurposed to reversibly inhibit the in vivo endocytosis of membrane proteins targeted by therapeutic monoclonal antibodies, as directly demonstrated by our human tumor ex vivo assay. Temporary endocytosis inhibition results in enhanced target availability and improved efficiency of natural killer cell-mediated antibody-dependent cellular cytotoxicity (ADCC), a mediator of clinical responses induced by IgG1 antibodies, demonstrated here for cetuximab, trastuzumab, and avelumab. Extensive analysis of downstream signaling pathways ruled out on-target toxicities. By overcoming the heterogeneity of drug target availability that frequently characterizes poorly responsive or resistant tumors, clinical application of reversible endocytosis inhibition may considerably improve the clinical benefit of ADCC-mediating therapeutic antibodies.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Drug Resistance, Neoplasm/immunology , Neoplasms/drug therapy , Prochlorperazine/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Antigen Presentation/drug effects , Biopsy , Cetuximab/pharmacology , Drug Delivery Systems/methods , Drug Resistance, Neoplasm/genetics , Endocytosis/drug effects , Endocytosis/immunology , Heterografts , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , MCF-7 Cells , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Neoplasms/genetics , Neoplasms/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Trastuzumab/pharmacology
8.
Annu Rev Cell Dev Biol ; 37: 115-142, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34242059

ABSTRACT

Microbes gain access to eukaryotic cells as food for bacteria-grazing protists, for host protection by microbe-killing immune cells, or for microbial benefit when pathogens enter host cells to replicate. But microbes can also gain access to a host cell and become an important-often required-beneficial partner. The oldest beneficial microbial infections are the ancient eukaryotic organelles now called the mitochondrion and plastid. But numerous other host-beneficial intracellular infections occur throughout eukaryotes. Here I review the genomics and cell biology of these interactions with a focus on intracellular bacteria. The genomes of host-beneficial intracellular bacteria have features that span a previously unfilled gap between pathogens and organelles. Host cell adaptations to allow the intracellular persistence of beneficial bacteria are found along with evidence for the microbial manipulation of host cells, but the cellular mechanisms of beneficial bacterial infections are not well understood.


Subject(s)
Bacteria , Organelles , Bacteria/genetics , Eukaryota , Eukaryotic Cells , Genomics , Host-Pathogen Interactions/genetics
9.
Annu Rev Cell Dev Biol ; 37: 369-389, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34196570

ABSTRACT

Wnt signaling has multiple functions beyond the transcriptional effects of ß-catenin stabilization. We review recent investigations that uncover new cell physiological effects through the regulation of Wnt receptor endocytosis, Wnt-induced stabilization of proteins (Wnt-STOP), macropinocytosis, increase in lysosomal activity, and metabolic changes. Many of these growth-promoting effects of canonical Wnt occur within minutes and are independent of new protein synthesis. A key element is the sequestration of glycogen synthase kinase 3 (GSK3) inside multivesicular bodies and lysosomes. Twenty percent of human proteins contain consecutive GSK3 phosphorylation motifs, which in the absence of Wnt can form phosphodegrons for polyubiquitination and proteasomal degradation. Wnt signaling by either the pharmacological inhibition of GSK3 or the loss of tumor-suppressor proteins, such as adenomatous polyposis coli (APC) and Axin1, increases lysosomal acidification, anabolic metabolites, and macropinocytosis, which is normally repressed by the GSK3-Axin1-APC destruction complex. The combination of these cell physiological effects drives cell growth.


Subject(s)
Glycogen Synthase Kinase 3 , Wnt Signaling Pathway , Glycogen Synthase Kinase 3/metabolism , Humans , Lysosomes/metabolism , Phosphorylation , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology
10.
Annu Rev Biochem ; 88: 461-485, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31220974

ABSTRACT

Glycosphingolipids are cell-type-specific components of the outer leaflet of mammalian plasma membranes. Gangliosides, sialic acid-containing glycosphingolipids, are especially enriched on neuronal surfaces. As amphi-philic molecules, they comprise a hydrophilic oligosaccharide chain attached to a hydrophobic membrane anchor, ceramide. Whereas glycosphingolipid formation is catalyzed by membrane-bound enzymes along the secretory pathway, degradation takes place at the surface of intralysosomal vesicles of late endosomes and lysosomes catalyzed in a stepwise fashion by soluble hydrolases and assisted by small lipid-binding glycoproteins. Inherited defects of lysosomal hydrolases or lipid-binding proteins cause the accumulation of undegradable material in lysosomal storage diseases (GM1 and GM2 gangliosidosis; Fabry, Gaucher, and Krabbe diseases; and metachromatic leukodystrophy). The catabolic processes are strongly modified by the lipid composition of the substrate-carrying membranes, and the pathological accumulation of primary storage compounds can trigger an accumulation of secondary storage compounds (e.g., small glycosphingolipids and cholesterol in Niemann-Pick disease).


Subject(s)
Glycosphingolipids , Lysosomal Storage Diseases/metabolism , Animals , Humans , Lysosomes/metabolism
11.
Cell ; 178(3): 536-551.e14, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31257024

ABSTRACT

The expression of some proteins in the autophagy pathway declines with age, which may impact neurodegeneration in diseases, including Alzheimer's Disease. We have identified a novel non-canonical function of several autophagy proteins in the conjugation of LC3 to Rab5+, clathrin+ endosomes containing ß-amyloid in a process of LC3-associated endocytosis (LANDO). We found that LANDO in microglia is a critical regulator of immune-mediated aggregate removal and microglial activation in a murine model of AD. Mice lacking LANDO but not canonical autophagy in the myeloid compartment or specifically in microglia have a robust increase in pro-inflammatory cytokine production in the hippocampus and increased levels of neurotoxic ß-amyloid. This inflammation and ß-amyloid deposition were associated with reactive microgliosis and tau hyperphosphorylation. LANDO-deficient AD mice displayed accelerated neurodegeneration, impaired neuronal signaling, and memory deficits. Our data support a protective role for LANDO in microglia in neurodegenerative pathologies resulting from ß-amyloid deposition.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Endocytosis , Microtubule-Associated Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Autophagy-Related Protein 5/deficiency , Autophagy-Related Protein 5/genetics , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , CD36 Antigens/metabolism , Cytokines/metabolism , Disease Models, Animal , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Microglia/cytology , Microglia/metabolism , RAW 264.7 Cells , Receptors, Immunologic/metabolism , Toll-Like Receptor 4/metabolism
12.
Cell ; 174(4): 884-896.e17, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30057119

ABSTRACT

Clathrin-mediated endocytosis is an essential cellular function in all eukaryotes that is driven by a self-assembled macromolecular machine of over 50 different proteins in tens to hundreds of copies. How these proteins are organized to produce endocytic vesicles with high precision and efficiency is not understood. Here, we developed high-throughput superresolution microscopy to reconstruct the nanoscale structural organization of 23 endocytic proteins from over 100,000 endocytic sites in yeast. We found that proteins assemble by radially ordered recruitment according to function. WASP family proteins form a circular nanoscale template on the membrane to spatially control actin nucleation during vesicle formation. Mathematical modeling of actin polymerization showed that this WASP nano-template optimizes force generation for membrane invagination and substantially increases the efficiency of endocytosis. Such nanoscale pre-patterning of actin nucleation may represent a general design principle for directional force generation in membrane remodeling processes such as during cell migration and division.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Endocytosis/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Secretory Vesicles/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , Actins/chemistry , Cell Membrane/metabolism , Microscopy, Fluorescence , Models, Theoretical , Protein Conformation , Wiskott-Aldrich Syndrome Protein Family/chemistry
13.
Cell ; 173(4): 934-945.e12, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29606354

ABSTRACT

Fusion is thought to open a pore to release vesicular cargoes vital for many biological processes, including exocytosis, intracellular trafficking, fertilization, and viral entry. However, fusion pores have not been observed and thus proved in live cells. Its regulatory mechanisms and functions remain poorly understood. With super-resolution STED microscopy, we observed dynamic fusion pore behaviors in live (neuroendocrine) cells, including opening, expansion, constriction, and closure, where pore size may vary between 0 and 490 nm within 26 milliseconds to seconds (vesicle size: 180-720 nm). These pore dynamics crucially determine the efficiency of vesicular cargo release and vesicle retrieval. They are generated by competition between pore expansion and constriction. Pharmacology and mutation experiments suggest that expansion and constriction are mediated by F-actin-dependent membrane tension and calcium/dynamin, respectively. These findings provide the missing live-cell evidence, proving the fusion-pore hypothesis, and establish a live-cell dynamic-pore theory accounting for fusion, fission, and their regulation.


Subject(s)
Cell Membrane/metabolism , Endocytosis/physiology , Membrane Fusion/physiology , Actins/metabolism , Animals , Calcium/metabolism , Cattle , Cell Membrane/chemistry , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , Dynamins/metabolism , Electric Stimulation , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Male , Microscopy, Confocal , Models, Biological , Patch-Clamp Techniques , Secretory Vesicles/physiology
14.
Cell ; 174(2): 325-337.e14, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29887380

ABSTRACT

Multiple proteins act co-operatively in mammalian clathrin-mediated endocytosis (CME) to generate endocytic vesicles from the plasma membrane. The principles controlling the activation and organization of the actin cytoskeleton during mammalian CME are, however, not fully understood. Here, we show that the protein FCHSD2 is a major activator of actin polymerization during CME. FCHSD2 deletion leads to decreased ligand uptake caused by slowed pit maturation. FCHSD2 is recruited to endocytic pits by the scaffold protein intersectin via an unusual SH3-SH3 interaction. Here, its flat F-BAR domain binds to the planar region of the plasma membrane surrounding the developing pit forming an annulus. When bound to the membrane, FCHSD2 activates actin polymerization by a mechanism that combines oligomerization and recruitment of N-WASP to PI(4,5)P2, thus promoting pit maturation. Our data therefore describe a molecular mechanism for linking spatiotemporally the plasma membrane to a force-generating actin platform guiding endocytic vesicle maturation.


Subject(s)
Actin Cytoskeleton/physiology , Carrier Proteins/metabolism , Clathrin/metabolism , Membrane Proteins/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/metabolism , Clathrin-Coated Vesicles/metabolism , Endocytosis , HeLa Cells , Humans , Liposomes/chemistry , Liposomes/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Microscopy, Fluorescence , Models, Molecular , Mutagenesis, Site-Directed , RNA Interference , RNA, Small Interfering/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/chemistry , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , src Homology Domains
15.
Annu Rev Cell Dev Biol ; 35: 55-84, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31283376

ABSTRACT

Endocytosis has long been identified as a key cellular process involved in bringing in nutrients, in clearing cellular debris in tissue, in the regulation of signaling, and in maintaining cell membrane compositional homeostasis. While clathrin-mediated endocytosis has been most extensively studied, a number of clathrin-independent endocytic pathways are continuing to be delineated. Here we provide a current survey of the different types of endocytic pathways available at the cell surface and discuss a new classification and plausible molecular mechanisms for some of the less characterized pathways. Along with an evolutionary perspective of the origins of some of these pathways, we provide an appreciation of the distinct roles that these pathways play in various aspects of cellular physiology, including the control of signaling and membrane tension.


Subject(s)
Cell Membrane/metabolism , Endocytosis , Signal Transduction , Animals , Biological Evolution , Cell Membrane/chemistry , Homeostasis , Humans
16.
Immunity ; 56(6): 1187-1203.e12, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37160118

ABSTRACT

B7 ligands (CD80 and CD86), expressed by professional antigen-presenting cells (APCs), activate the main co-stimulatory receptor CD28 on T cells in trans. However, in peripheral tissues, APCs expressing B7 ligands are relatively scarce. This raises the questions of whether and how CD28 co-stimulation occurs in peripheral tissues. Here, we report that CD8+ T cells displayed B7 ligands that interacted with CD28 in cis at membrane invaginations of the immunological synapse as a result of membrane remodeling driven by phosphoinositide-3-kinase (PI3K) and sorting-nexin-9 (SNX9). cis-B7:CD28 interactions triggered CD28 signaling through protein kinase C theta (PKCθ) and promoted CD8+ T cell survival, migration, and cytokine production. In mouse tumor models, loss of T cell-intrinsic cis-B7:CD28 interactions decreased intratumoral T cells and accelerated tumor growth. Thus, B7 ligands on CD8+ T cells can evoke cell-autonomous CD28 co-stimulation in cis in peripheral tissues, suggesting cis-signaling as a general mechanism for boosting T cell functionality.


Subject(s)
CD28 Antigens , CD8-Positive T-Lymphocytes , Mice , Animals , CD28 Antigens/metabolism , Antigens, CD/metabolism , Ligands , Synaptic Membranes/metabolism , B7-2 Antigen , Membrane Glycoproteins/metabolism , B7-1 Antigen/metabolism , Cell Adhesion Molecules , Lymphocyte Activation
17.
Cell ; 171(6): 1383-1396.e12, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29195077

ABSTRACT

DSL ligands activate Notch by inducing proteolytic cleavage of the receptor ectodomain, an event that requires ligand to be endocytosed in signal-sending cells by the adaptor protein Epsin. Two classes of explanation for this unusual requirement are (1) recycling models, in which the ligand must be endocytosed to be modified or repositioned before it binds Notch and (2) pulling models, in which the ligand must be endocytosed after it binds Notch to exert force that exposes an otherwise buried site for cleavage. We demonstrate in vivo that ligands that cannot enter the Epsin pathway nevertheless bind Notch but fail to activate the receptor because they cannot exert sufficient force. This argues against recycling models and in favor of pulling models. Our results also suggest that once ligand binds receptor, activation depends on a competition between Epsin-mediated ligand endocytosis, which induces cleavage, and transendocytosis of the ligand by the receptor, which aborts the incipient signal.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/cytology , Drosophila/metabolism , Endocytosis , Signal Transduction , Vesicular Transport Proteins/metabolism , Wings, Animal/metabolism , Animals , Drosophila/growth & development , Imaginal Discs/metabolism , Ligands , Receptors, Notch/metabolism
18.
Cell ; 170(1): 172-184.e11, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28648660

ABSTRACT

Membrane scission is essential for intracellular trafficking. While BAR domain proteins such as endophilin have been reported in dynamin-independent scission of tubular membrane necks, the cutting mechanism has yet to be deciphered. Here, we combine a theoretical model, in vitro, and in vivo experiments revealing how protein scaffolds may cut tubular membranes. We demonstrate that the protein scaffold bound to the underlying tube creates a frictional barrier for lipid diffusion; tube elongation thus builds local membrane tension until the membrane undergoes scission through lysis. We call this mechanism friction-driven scission (FDS). In cells, motors pull tubes, particularly during endocytosis. Through reconstitution, we show that motors not only can pull out and extend protein-scaffolded tubes but also can cut them by FDS. FDS is generic, operating even in the absence of amphipathic helices in the BAR domain, and could in principle apply to any high-friction protein and membrane assembly.


Subject(s)
Endocytosis , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Acyltransferases/chemistry , Acyltransferases/metabolism , Animals , Biomechanical Phenomena , Friction , Humans , Lipid Metabolism , Protein Domains , Rats
19.
Annu Rev Cell Dev Biol ; 34: 111-136, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30296391

ABSTRACT

The plasma membrane of eukaryotic cells is not a simple sheet of lipids and proteins but is differentiated into subdomains with crucial functions. Caveolae, small pits in the plasma membrane, are the most abundant surface subdomains of many mammalian cells. The cellular functions of caveolae have long remained obscure, but a new molecular understanding of caveola formation has led to insights into their workings. Caveolae are formed by the coordinated action of a number of lipid-interacting proteins to produce a microdomain with a specific structure and lipid composition. Caveolae can bud from the plasma membrane to form an endocytic vesicle or can flatten into the membrane to help cells withstand mechanical stress. The role of caveolae as mechanoprotective and signal transduction elements is reviewed in the context of disease conditions associated with caveola dysfunction.


Subject(s)
Caveolae/metabolism , Cell Membrane/genetics , Transport Vesicles/genetics , Caveolae/chemistry , Caveolae/pathology , Cell Membrane/chemistry , Endocytosis/genetics , Humans , Signal Transduction/genetics , Stress, Mechanical , Structure-Activity Relationship , Transport Vesicles/chemistry
20.
Mol Cell ; 84(2): 386-400.e11, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38103558

ABSTRACT

The posttranslational modifier ubiquitin regulates most cellular processes. Its ability to form polymeric chains of distinct linkages is key to its diverse functionality. Yet, we still lack the experimental tools to induce linkage-specific polyubiquitylation of a protein of interest in cells. Here, we introduce a set of engineered ubiquitin protein ligases and matching ubiquitin acceptor tags for the rapid, inducible linear (M1-), K48-, or K63-linked polyubiquitylation of proteins in yeast and mammalian cells. By applying the so-called "Ubiquiton" system to proteasomal targeting and the endocytic pathway, we validate this tool for soluble cytoplasmic and nuclear as well as chromatin-associated and integral membrane proteins and demonstrate how it can be used to control the localization and stability of its targets. We expect that the Ubiquiton system will serve as a versatile, broadly applicable research tool to explore the signaling functions of polyubiquitin chains in many biological contexts.


Subject(s)
Ubiquitin-Protein Ligases , Ubiquitin , Animals , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Polyubiquitin/genetics , Polyubiquitin/metabolism , Signal Transduction , Proteasome Endopeptidase Complex/metabolism , Ubiquitination , Mammals/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL