Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.487
Filter
Add more filters

Publication year range
1.
Cell ; 186(21): 4710-4727.e35, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37774705

ABSTRACT

Polarized cells rely on a polarized cytoskeleton to function. Yet, how cortical polarity cues induce cytoskeleton polarization remains elusive. Here, we capitalized on recently established designed 2D protein arrays to ectopically engineer cortical polarity of virtually any protein of interest during mitosis in various cell types. This enables direct manipulation of polarity signaling and the identification of the cortical cues sufficient for cytoskeleton polarization. Using this assay, we dissected the logic of the Par complex pathway, a key regulator of cytoskeleton polarity during asymmetric cell division. We show that cortical clustering of any Par complex subunit is sufficient to trigger complex assembly and that the primary kinetic barrier to complex assembly is the relief of Par6 autoinhibition. Further, we found that inducing cortical Par complex polarity induces two hallmarks of asymmetric cell division in unpolarized mammalian cells: spindle orientation, occurring via Par3, and central spindle asymmetry, depending on aPKC activity.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Polarity , Cytological Techniques , Mitosis , Animals , Cytoskeleton/metabolism , Mammals/metabolism , Microtubules/metabolism , Protein Kinase C/metabolism , Adaptor Proteins, Signal Transducing/metabolism
2.
Cell ; 186(14): 3049-3061.e15, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37311454

ABSTRACT

Membrane tension is thought to be a long-range integrator of cell physiology. Membrane tension has been proposed to enable cell polarity during migration through front-back coordination and long-range protrusion competition. These roles necessitate effective tension transmission across the cell. However, conflicting observations have left the field divided as to whether cell membranes support or resist tension propagation. This discrepancy likely originates from the use of exogenous forces that may not accurately mimic endogenous forces. We overcome this complication by leveraging optogenetics to directly control localized actin-based protrusions or actomyosin contractions while simultaneously monitoring the propagation of membrane tension using dual-trap optical tweezers. Surprisingly, actin-driven protrusions and actomyosin contractions both elicit rapid global membrane tension propagation, whereas forces applied to cell membranes alone do not. We present a simple unifying mechanical model in which mechanical forces that engage the actin cortex drive rapid, robust membrane tension propagation through long-range membrane flows.


Subject(s)
Actins , Actomyosin , Actins/metabolism , Actomyosin/metabolism , Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Cell Movement/physiology
3.
Cell ; 186(25): 5656-5672.e21, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38029746

ABSTRACT

Molecular signals interact in networks to mediate biological processes. To analyze these networks, it would be useful to image many signals at once, in the same living cell, using standard microscopes and genetically encoded fluorescent reporters. Here, we report temporally multiplexed imaging (TMI), which uses genetically encoded fluorescent proteins with different clocklike properties-such as reversibly photoswitchable fluorescent proteins with different switching kinetics-to represent different cellular signals. We linearly decompose a brief (few-second-long) trace of the fluorescence fluctuations, at each point in a cell, into a weighted sum of the traces exhibited by each fluorophore expressed in the cell. The weights then represent the signal amplitudes. We use TMI to analyze relationships between different kinase activities in individual cells, as well as between different cell-cycle signals, pointing toward broad utility throughout biology in the analysis of signal transduction cascades in living systems.


Subject(s)
Proteins , Signal Transduction , Animals , Humans , Mice , Cell Line , Fluorescent Dyes , Microscopy, Fluorescence/methods , Phosphorylation , Cell Survival
4.
Cell ; 185(8): 1356-1372.e26, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35395179

ABSTRACT

Tumor-resident intracellular microbiota is an emerging tumor component that has been documented for a variety of cancer types with unclear biological functions. Here, we explored the functional significance of these intratumor bacteria, primarily using a murine spontaneous breast-tumor model MMTV-PyMT. We found that depletion of intratumor bacteria significantly reduced lung metastasis without affecting primary tumor growth. During metastatic colonization, intratumor bacteria carried by circulating tumor cells promoted host-cell survival by enhancing resistance to fluid shear stress by reorganizing actin cytoskeleton. We further showed that intratumor administration of selected bacteria strains isolated from tumor-resident microbiota promoted metastasis in two murine tumor models with significantly different levels of metastasis potential. Our findings suggest that tumor-resident microbiota, albeit at low biomass, play an important role in promoting cancer metastasis, intervention of which might therefore be worth exploring for advancing oncology care.


Subject(s)
Breast Neoplasms , Microbiota , Neoplasm Metastasis , Animals , Breast Neoplasms/microbiology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/pathology , Mice , Neoplastic Cells, Circulating/pathology
5.
Cell ; 185(19): 3588-3602.e21, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36113429

ABSTRACT

The current dogma of RNA-mediated innate immunity is that sensing of immunostimulatory RNA ligands is sufficient for the activation of intracellular sensors and induction of interferon (IFN) responses. Here, we report that actin cytoskeleton disturbance primes RIG-I-like receptor (RLR) activation. Actin cytoskeleton rearrangement induced by virus infection or commonly used reagents to intracellularly deliver RNA triggers the relocalization of PPP1R12C, a regulatory subunit of the protein phosphatase-1 (PP1), from filamentous actin to cytoplasmic RLRs. This allows dephosphorylation-mediated RLR priming and, together with the RNA agonist, induces effective RLR downstream signaling. Genetic ablation of PPP1R12C impairs antiviral responses and enhances susceptibility to infection with several RNA viruses including SARS-CoV-2, influenza virus, picornavirus, and vesicular stomatitis virus. Our work identifies actin cytoskeleton disturbance as a priming signal for RLR-mediated innate immunity, which may open avenues for antiviral or adjuvant design.


Subject(s)
Actins , COVID-19 , Actin Cytoskeleton , Antiviral Agents , Humans , Interferons , Ligands , Protein Phosphatase 1 , RNA , RNA Helicases , Receptors, Retinoic Acid/metabolism , SARS-CoV-2
6.
Cell ; 185(23): 4376-4393.e18, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36318920

ABSTRACT

The function of biomolecular condensates is often restricted by condensate dissolution. Whether condensates can be suppressed without condensate dissolution is unclear. Here, we show that upstream regulators of the Hippo signaling pathway form functionally antagonizing condensates, and their coalescence into a common phase provides a mode of counteracting the function of biomolecular condensates without condensate dissolution. Specifically, the negative regulator SLMAP forms Hippo-inactivating condensates to facilitate pathway inhibition by the STRIPAK complex. In response to cell-cell contact or osmotic stress, the positive regulators AMOT and KIBRA form Hippo-activating condensates to facilitate pathway activation. The functionally antagonizing SLMAP and AMOT/KIBRA condensates further coalesce into a common phase to inhibit STRIPAK function. These findings provide a paradigm for restricting the activity of biomolecular condensates without condensate dissolution, shed light on the molecular principles of multiphase organization, and offer a conceptual framework for understanding upstream regulation of the Hippo signaling pathway.


Subject(s)
Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Signal Transduction
7.
Cell ; 184(14): 3660-3673.e18, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34166615

ABSTRACT

Membrane remodeling and repair are essential for all cells. Proteins that perform these functions include Vipp1/IM30 in photosynthetic plastids, PspA in bacteria, and ESCRT-III in eukaryotes. Here, using a combination of evolutionary and structural analyses, we show that these protein families are homologous and share a common ancient evolutionary origin that likely predates the last universal common ancestor. This homology is evident in cryo-electron microscopy structures of Vipp1 rings from the cyanobacterium Nostoc punctiforme presented over a range of symmetries. Each ring is assembled from rungs that stack and progressively tilt to form dome-shaped curvature. Assembly is facilitated by hinges in the Vipp1 monomer, similar to those in ESCRT-III proteins, which allow the formation of flexible polymers. Rings have an inner lumen that is able to bind and deform membranes. Collectively, these data suggest conserved mechanistic principles that underlie Vipp1, PspA, and ESCRT-III-dependent membrane remodeling across all domains of life.


Subject(s)
Bacterial Proteins/metabolism , Cell Membrane/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Heat-Shock Proteins/metabolism , Multigene Family , Nostoc/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/ultrastructure , Chickens , Cryoelectron Microscopy , Endosomal Sorting Complexes Required for Transport/chemistry , Evolution, Molecular , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/ultrastructure , Humans , Models, Molecular , Protein Structure, Secondary , Sequence Homology, Amino Acid , Thermodynamics
8.
Cell ; 184(20): 5107-5121.e14, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34551316

ABSTRACT

Neural circuit assembly features simultaneous targeting of numerous neuronal processes from constituent neuron types, yet the dynamics is poorly understood. Here, we use the Drosophila olfactory circuit to investigate dynamic cellular processes by which olfactory receptor neurons (ORNs) target axons precisely to specific glomeruli in the ipsi- and contralateral antennal lobes. Time-lapse imaging of individual axons from 30 ORN types revealed a rich diversity in extension speed, innervation timing, and ipsilateral branch locations and identified that ipsilateral targeting occurs via stabilization of transient interstitial branches. Fast imaging using adaptive optics-corrected lattice light-sheet microscopy showed that upon approaching target, many ORN types exhibiting "exploring branches" consisted of parallel microtubule-based terminal branches emanating from an F-actin-rich hub. Antennal nerve ablations uncovered essential roles for bilateral axons in contralateral target selection and for ORN axons to facilitate dendritic refinement of postsynaptic partner neurons. Altogether, these observations provide cellular bases for wiring specificity establishment.


Subject(s)
Olfactory Pathways/cytology , Olfactory Pathways/diagnostic imaging , Time-Lapse Imaging , Animals , Axons/physiology , Cells, Cultured , Dendrites/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Microtubules/metabolism , Olfactory Receptor Neurons/physiology , Time Factors
9.
Annu Rev Cell Dev Biol ; 38: 155-178, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35905769

ABSTRACT

Eukaryotic cells across the tree of life organize their subcellular components via intracellular transport mechanisms. In canonical transport, myosin, kinesin, and dynein motor proteins interact with cargos via adaptor proteins and move along filamentous actin or microtubule tracks. In contrast to this canonical mode, hitchhiking is a newly discovered mode of intracellular transport in which a cargo attaches itself to an already-motile cargo rather than directly associating with a motor protein itself. Many cargos including messenger RNAs, protein complexes, and organelles hitchhike on membrane-bound cargos. Hitchhiking-like behaviors have been shown to impact cellular processes including local protein translation, long-distance signaling, and organelle network reorganization. Here, we review instances of cargo hitchhiking in fungal, animal, and plant cells and discuss the potential cellular and evolutionary importance of hitchhiking in these different contexts.


Subject(s)
Dyneins , Kinesins , Actins/metabolism , Animals , Dyneins/genetics , Dyneins/metabolism , Kinesins/genetics , Microtubules/genetics , Microtubules/metabolism , Myosins/genetics , Myosins/metabolism , Plant Cells/metabolism
10.
Cell ; 180(3): 568-584.e23, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31981491

ABSTRACT

We present the largest exome sequencing study of autism spectrum disorder (ASD) to date (n = 35,584 total samples, 11,986 with ASD). Using an enhanced analytical framework to integrate de novo and case-control rare variation, we identify 102 risk genes at a false discovery rate of 0.1 or less. Of these genes, 49 show higher frequencies of disruptive de novo variants in individuals ascertained to have severe neurodevelopmental delay, whereas 53 show higher frequencies in individuals ascertained to have ASD; comparing ASD cases with mutations in these groups reveals phenotypic differences. Expressed early in brain development, most risk genes have roles in regulation of gene expression or neuronal communication (i.e., mutations effect neurodevelopmental and neurophysiological changes), and 13 fall within loci recurrently hit by copy number variants. In cells from the human cortex, expression of risk genes is enriched in excitatory and inhibitory neuronal lineages, consistent with multiple paths to an excitatory-inhibitory imbalance underlying ASD.


Subject(s)
Autistic Disorder/genetics , Cerebral Cortex/growth & development , Exome Sequencing/methods , Gene Expression Regulation, Developmental , Neurobiology/methods , Case-Control Studies , Cell Lineage , Cohort Studies , Exome , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Mutation, Missense , Neurons/metabolism , Phenotype , Sex Factors , Single-Cell Analysis/methods
11.
Annu Rev Cell Dev Biol ; 37: 23-41, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34186005

ABSTRACT

The purpose of this review is to explore self-organizing mechanisms that pattern microtubules (MTs) and spatially organize animal cell cytoplasm, inspired by recent experiments in frog egg extract. We start by reviewing conceptual distinctions between self-organizing and templating mechanisms for subcellular organization. We then discuss self-organizing mechanisms that generate radial MT arrays and cell centers in the absence of centrosomes. These include autocatalytic MT nucleation, transport of minus ends, and nucleation from organelles such as melanosomes and Golgi vesicles that are also dynein cargoes. We then discuss mechanisms that partition the cytoplasm in syncytia, in which multiple nuclei share a common cytoplasm, starting with cytokinesis, when all metazoan cells are transiently syncytial. The cytoplasm of frog eggs is partitioned prior to cytokinesis by two self-organizing modules, protein regulator of cytokinesis 1 (PRC1)-kinesin family member 4A (KIF4A) and chromosome passenger complex (CPC)-KIF20A. Similar modules may partition longer-lasting syncytia, such as early Drosophila embryos. We end by discussing shared mechanisms and principles for the MT-based self-organization of cellular units.


Subject(s)
Centrosome , Microtubules , Animals , Centrosome/metabolism , Cytokinesis , Cytoskeleton , Golgi Apparatus , Microtubules/metabolism
12.
Annu Rev Cell Dev Biol ; 37: 1-21, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34186006

ABSTRACT

One of the most common bacterial shapes is a rod, yet we have a limited understanding of how this simple shape is constructed. While only six proteins are required for rod shape, we are just beginning to understand how they self-organize to build the micron-sized enveloping structures that define bacterial shape out of nanometer-sized glycan strains. Here, we detail and summarize the insights gained over the last 20 years into this complex problem that have been achieved with a wide variety of different approaches. We also explain and compare both current and past models of rod shape formation and maintenance and then highlight recent insights into how the Rod complex might be regulated.


Subject(s)
Bacteria , Bacterial Proteins , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics
13.
Cell ; 179(1): 120-131.e13, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31539492

ABSTRACT

Focal adhesions (FAs) are protein machineries essential for cell adhesion, migration, and differentiation. Talin is an integrin-activating and tension-sensing FA component directly connecting integrins in the plasma membrane with the actomyosin cytoskeleton. To understand how talin function is regulated, we determined a cryoelectron microscopy (cryo-EM) structure of full-length talin1 revealing a two-way mode of autoinhibition. The actin-binding rod domains fold into a 15-nm globular arrangement that is interlocked by the integrin-binding FERM head. In turn, the rod domains R9 and R12 shield access of the FERM domain to integrin and the phospholipid PIP2 at the membrane. This mechanism likely ensures synchronous inhibition of integrin, membrane, and cytoskeleton binding. We also demonstrate that compacted talin1 reversibly unfolds to an ∼60-nm string-like conformation, revealing interaction sites for vinculin and actin. Our data explain how fast switching between active and inactive conformations of talin could regulate FA turnover, a process critical for cell adhesion and signaling.


Subject(s)
Focal Adhesions/metabolism , Protein Interaction Domains and Motifs , Talin/chemistry , Talin/metabolism , Actins/metabolism , Actomyosin/metabolism , Binding Sites , Cell Adhesion/physiology , Cryoelectron Microscopy , Cytoskeleton/metabolism , Dimerization , Escherichia coli/metabolism , Humans , Integrins/metabolism , Models, Molecular , Protein Binding , Signal Transduction/physiology , Vinculin/metabolism
14.
Cell ; 176(1-2): 73-84.e15, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30612742

ABSTRACT

Local translation meets protein turnover and plasticity demands at synapses, however, the location of its energy supply is unknown. We found that local translation in neurons is powered by mitochondria and not by glycolysis. Super-resolution microscopy revealed that dendritic mitochondria exist as stable compartments of single or multiple filaments. To test if these mitochondrial compartments can serve as local energy supply for synaptic translation, we stimulated individual synapses to induce morphological plasticity and visualized newly synthesized proteins. Depletion of local mitochondrial compartments abolished both the plasticity and the stimulus-induced synaptic translation. These mitochondrial compartments serve as spatially confined energy reserves, as local depletion of a mitochondrial compartment did not affect synaptic translation at remote spines. The length and stability of dendritic mitochondrial compartments and the spatial functional domain were altered by cytoskeletal disruption. These results indicate that cytoskeletally tethered local energy compartments exist in dendrites to fuel local translation during synaptic plasticity.


Subject(s)
Mitochondria/metabolism , Neurons/metabolism , Protein Biosynthesis/physiology , Animals , Cytoskeleton/metabolism , Dendrites/metabolism , Dendritic Spines/metabolism , Female , Male , Mitochondria/physiology , Neuronal Plasticity/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Synapses/metabolism
15.
Cell ; 179(7): 1512-1524.e15, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31835030

ABSTRACT

During cell division, newly replicated DNA is actively segregated to the daughter cells. In most bacteria, this process involves the DNA-binding protein ParB, which condenses the centromeric regions of sister DNA molecules into kinetochore-like structures that recruit the DNA partition ATPase ParA and the prokaroytic SMC/condensin complex. Here, we report the crystal structure of a ParB-like protein (PadC) that emerges to tightly bind the ribonucleotide CTP. The CTP-binding pocket of PadC is conserved in ParB and composed of signature motifs known to be essential for ParB function. We find that ParB indeed interacts with CTP and requires nucleotide binding for DNA condensation in vivo. We further show that CTP-binding modulates the affinity of ParB for centromeric parS sites, whereas parS recognition stimulates its CTPase activity. ParB proteins thus emerge as a new class of CTP-dependent molecular switches that act in concert with ATPases and GTPases to control fundamental cellular functions.


Subject(s)
Bacterial Proteins/chemistry , Cytidine Triphosphate/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Myxococcus xanthus/genetics , Myxococcus xanthus/metabolism , Nucleotide Motifs , Protein Binding
16.
Annu Rev Cell Dev Biol ; 36: 191-218, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32663035

ABSTRACT

Neutrophils are critical to innate immunity, including host defense against bacterial and fungal infections. They achieve their host defense role by phagocytosing pathogens, secreting their granules full of cytotoxic enzymes, or expelling neutrophil extracellular traps (NETs) during the process of NETosis. NETs are weblike DNA structures decorated with histones and antimicrobial proteins released by activated neutrophils. Initially described as a means for neutrophils to neutralize pathogens, NET release also occurs in sterile inflammation, promotes thrombosis, and can mediate tissue damage. To effectively manipulate this double-edged sword to fight a particular disease, researchers must work toward understanding the mechanisms driving NETosis. Such understanding would allow the generation of new drugs to promote or prevent NETosis as needed. While knowledge regarding the (patho)physiological roles of NETosis is accumulating, little is known about the cellular and biophysical bases of this process. In this review, we describe and discuss our current knowledge of the molecular, cellular, and biophysical mechanisms mediating NET release as well as open questions in the field.


Subject(s)
Extracellular Traps/metabolism , Animals , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Cytosol/metabolism , DNA/metabolism , Humans
17.
Annu Rev Biochem ; 87: 839-869, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29494237

ABSTRACT

Cells depend on hugely diverse lipidomes for many functions. The actions and structural integrity of the plasma membrane and most organelles also critically depend on membranes and their lipid components. Despite the biological importance of lipids, our understanding of lipid engagement, especially the roles of lipid hydrophobic alkyl side chains, in key cellular processes is still developing. Emerging research has begun to dissect the importance of lipids in intricate events such as cell division. This review discusses how these structurally diverse biomolecules are spatially and temporally regulated during cell division, with a focus on cytokinesis. We analyze how lipids facilitate changes in cellular morphology during division and how they participate in key signaling events. We identify which cytokinesis proteins are associated with membranes, suggesting lipid interactions. More broadly, we highlight key unaddressed questions in lipid cell biology and techniques, including mass spectrometry, advanced imaging, and chemical biology, which will help us gain insights into the functional roles of lipids.


Subject(s)
Cell Division/physiology , Lipid Metabolism , Animals , Cell Cycle/physiology , Humans , Lipids/chemistry , Mass Spectrometry , Models, Biological , Models, Molecular , Molecular Structure , Signal Transduction
18.
Cell ; 172(5): 1063-1078.e19, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474907

ABSTRACT

Interneurons navigate along multiple tangential paths to settle into appropriate cortical layers. They undergo a saltatory migration paced by intermittent nuclear jumps whose regulation relies on interplay between extracellular cues and genetic-encoded information. It remains unclear how cycles of pause and movement are coordinated at the molecular level. Post-translational modification of proteins contributes to cell migration regulation. The present study uncovers that carboxypeptidase 1, which promotes post-translational protein deglutamylation, controls the pausing of migrating cortical interneurons. Moreover, we demonstrate that pausing during migration attenuates movement simultaneity at the population level, thereby controlling the flow of interneurons invading the cortex. Interfering with the regulation of pausing not only affects the size of the cortical interneuron cohort but also impairs the generation of age-matched projection neurons of the upper layers.


Subject(s)
Cell Movement , Cerebral Cortex/cytology , Interneurons/cytology , Morphogenesis , Actomyosin/metabolism , Animals , Carboxypeptidases/metabolism , Cell Cycle , Chemotactic Factors/metabolism , Embryo, Mammalian/cytology , Female , Gene Deletion , Interneurons/metabolism , Mice , Mice, Knockout , Myosin-Light-Chain Kinase/metabolism , Neurogenesis , Phenotype
19.
Cell ; 175(3): 796-808.e14, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30340043

ABSTRACT

During cell division, mitotic motors organize microtubules in the bipolar spindle into either polar arrays at the spindle poles or a "nematic" network of aligned microtubules at the spindle center. The reasons for the distinct self-organizing capacities of dynamic microtubules and different motors are not understood. Using in vitro reconstitution experiments and computer simulations, we show that the human mitotic motors kinesin-5 KIF11 and kinesin-14 HSET, despite opposite directionalities, can both organize dynamic microtubules into either polar or nematic networks. We show that in addition to the motor properties the natural asymmetry between microtubule plus- and minus-end growth critically contributes to the organizational potential of the motors. We identify two control parameters that capture system composition and kinetic properties and predict the outcome of microtubule network organization. These results elucidate a fundamental design principle of spindle bipolarity and establish general rules for active filament network organization.


Subject(s)
Kinesins/metabolism , Microtubules/metabolism , Molecular Dynamics Simulation , Spindle Apparatus/metabolism , Animals , Humans , Kinesins/chemistry , Microtubules/chemistry , Sf9 Cells , Spindle Apparatus/chemistry , Spodoptera
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL