Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38915582

ABSTRACT

Single-particle tracking demonstrates that individual filaments in bundles of vimentin intermediate filaments are transported in the cytoplasm by motor proteins along microtubules. Furthermore, using 3D FIB-SEM the authors showed that vimentin filament bundles are loosely packed and coaligned with microtubules. Vimentin intermediate filaments (VIFs) form complex, tight-packed networks; due to this density, traditional ensemble labeling and imaging approaches cannot accurately discern single filament behavior. To address this, we introduce a sparse vimentin-SunTag labeling strategy to unambiguously visualize individual filament dynamics. This technique confirmed known long-range dynein and kinesin transport of peripheral VIFs and uncovered extensive bidirectional VIF motion within the perinuclear vimentin network, a region we had thought too densely bundled to permit such motility. To examine the nanoscale organization of perinuclear vimentin, we acquired high-resolution electron microscopy volumes of a vitreously frozen cell and reconstructed VIFs and microtubules within a ~50 µm3 window. Of 583 VIFs identified, most were integrated into long, semi-coherent bundles that fluctuated in width and filament packing density. Unexpectedly, VIFs displayed minimal local co-alignment with microtubules, save for sporadic cross-over sites that we predict facilitate cytoskeletal crosstalk. Overall, this work demonstrates single VIF dynamics and organization in the cellular milieu for the first time.

2.
Dev Cell ; 59(11): 1410-1424.e4, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38593803

ABSTRACT

Endoplasmic reticulum exit sites (ERESs) are tubular outgrowths of endoplasmic reticulum that serve as the earliest station for protein sorting and export into the secretory pathway. How these structures respond to different cellular conditions remains unclear. Here, we report that ERESs undergo lysosome-dependent microautophagy when Ca2+ is released by lysosomes in response to nutrient stressors such as mTOR inhibition or amino acid starvation in mammalian cells. Targeting and uptake of ERESs into lysosomes were observed by super-resolution live-cell imaging and focus ion beam scanning electron microscopy (FIB-SEM). The mechanism was ESCRT dependent and required ubiquitinated SEC31, ALG2, and ALIX, with a knockout of ALG2 or function-blocking mutations of ALIX preventing engulfment of ERESs by lysosomes. In vitro, reconstitution of the pathway was possible using lysosomal lipid-mimicking giant unilamellar vesicles and purified recombinant components. Together, these findings demonstrate a pathway of lysosome-dependent ERES microautophagy mediated by COPII, ALG2, and ESCRTS induced by nutrient stress.


Subject(s)
COP-Coated Vesicles , Calcium-Binding Proteins , Endoplasmic Reticulum , Endosomal Sorting Complexes Required for Transport , Lysosomes , Microautophagy , Vesicular Transport Proteins , Lysosomes/metabolism , Endoplasmic Reticulum/metabolism , Humans , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , COP-Coated Vesicles/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Protein Transport , HeLa Cells , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Autophagy/physiology , TOR Serine-Threonine Kinases/metabolism , Calcium/metabolism
3.
Nature ; 626(7997): 169-176, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38267577

ABSTRACT

To coordinate cellular physiology, eukaryotic cells rely on the rapid exchange of molecules at specialized organelle-organelle contact sites1,2. Endoplasmic reticulum-mitochondrial contact sites (ERMCSs) are particularly vital communication hubs, playing key roles in the exchange of signalling molecules, lipids and metabolites3,4. ERMCSs are maintained by interactions between complementary tethering molecules on the surface of each organelle5,6. However, due to the extreme sensitivity of these membrane interfaces to experimental perturbation7,8, a clear understanding of their nanoscale organization and regulation is still lacking. Here we combine three-dimensional electron microscopy with high-speed molecular tracking of a model organelle tether, Vesicle-associated membrane protein (VAMP)-associated protein B (VAPB), to map the structure and diffusion landscape of ERMCSs. We uncovered dynamic subdomains within VAPB contact sites that correlate with ER membrane curvature and undergo rapid remodelling. We show that VAPB molecules enter and leave ERMCSs within seconds, despite the contact site itself remaining stable over much longer time scales. This metastability allows ERMCSs to remodel with changes in the physiological environment to accommodate metabolic needs of the cell. An amyotrophic lateral sclerosis-associated mutation in VAPB perturbs these subdomains, likely impairing their remodelling capacity and resulting in impaired interorganelle communication. These results establish high-speed single-molecule imaging as a new tool for mapping the structure of contact site interfaces and reveal that the diffusion landscape of VAPB at contact sites is a crucial component of ERMCS homeostasis.


Subject(s)
Endoplasmic Reticulum , Mitochondria , Mitochondrial Membranes , Movement , Vesicular Transport Proteins , Humans , Amyotrophic Lateral Sclerosis/genetics , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Mitochondria/chemistry , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Signal Transduction , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/ultrastructure , Microscopy, Electron , Imaging, Three-Dimensional , Binding Sites , Diffusion , Time Factors , Mutation , Homeostasis
4.
Science ; 376(6591): 377-382, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35446649

ABSTRACT

Cytotoxic T lymphocytes (CTLs) and natural killer cells kill virus-infected and tumor cells through the polarized release of perforin and granzymes. Perforin is a pore-forming toxin that creates a lesion in the plasma membrane of the target cell through which granzymes enter the cytosol and initiate apoptosis. Endosomal sorting complexes required for transport (ESCRT) proteins are involved in the repair of small membrane wounds. We found that ESCRT proteins were precisely recruited in target cells to sites of CTL engagement immediately after perforin release. Inhibition of ESCRT machinery in cancer-derived cells enhanced their susceptibility to CTL-mediated killing. Thus, repair of perforin pores by ESCRT machinery limits granzyme entry into the cytosol, potentially enabling target cells to resist cytolytic attack.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Membrane Glycoproteins , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Granzymes/metabolism , Membrane Glycoproteins/metabolism , Perforin/genetics , Perforin/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , T-Lymphocytes, Cytotoxic/metabolism
6.
Nature ; 599(7883): 147-151, 2021 11.
Article in English | MEDLINE | ID: mdl-34616045

ABSTRACT

Understanding cellular architecture is essential for understanding biology. Electron microscopy (EM) uniquely visualizes cellular structures with nanometre resolution. However, traditional methods, such as thin-section EM or EM tomography, have limitations in that they visualize only a single slice or a relatively small volume of the cell, respectively. Focused ion beam-scanning electron microscopy (FIB-SEM) has demonstrated the ability to image small volumes of cellular samples with 4-nm isotropic voxels1. Owing to advances in the precision and stability of FIB milling, together with enhanced signal detection and faster SEM scanning, we have increased the volume that can be imaged with 4-nm voxels by two orders of magnitude. Here we present a volume EM atlas at such resolution comprising ten three-dimensional datasets for whole cells and tissues, including cancer cells, immune cells, mouse pancreatic islets and Drosophila neural tissues. These open access data (via OpenOrganelle2) represent the foundation of a field of high-resolution whole-cell volume EM and subsequent analyses, and we invite researchers to explore this atlas and pose questions.


Subject(s)
Datasets as Topic , Information Dissemination , Microscopy, Electron, Scanning , Organelles/ultrastructure , Animals , Cell Line , Cells, Cultured , Drosophila melanogaster/cytology , Drosophila melanogaster/ultrastructure , Female , Golgi Apparatus/ultrastructure , Humans , Interphase , Islets of Langerhans/cytology , Male , Mice , Microscopy, Electron, Scanning/methods , Microscopy, Electron, Scanning/standards , Microtubules/ultrastructure , Neuroglia/ultrastructure , Neurons/ultrastructure , Open Access Publishing , Ovarian Neoplasms/immunology , Ovarian Neoplasms/ultrastructure , Ribosomes/ultrastructure , Synaptic Vesicles/ultrastructure , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure
7.
Cell ; 184(9): 2412-2429.e16, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33852913

ABSTRACT

Cellular versatility depends on accurate trafficking of diverse proteins to their organellar destinations. For the secretory pathway (followed by approximately 30% of all proteins), the physical nature of the vessel conducting the first portage (endoplasmic reticulum [ER] to Golgi apparatus) is unclear. We provide a dynamic 3D view of early secretory compartments in mammalian cells with isotropic resolution and precise protein localization using whole-cell, focused ion beam scanning electron microscopy with cryo-structured illumination microscopy and live-cell synchronized cargo release approaches. Rather than vesicles alone, the ER spawns an elaborate, interwoven tubular network of contiguous lipid bilayers (ER exit site) for protein export. This receptacle is capable of extending microns along microtubules while still connected to the ER by a thin neck. COPII localizes to this neck region and dynamically regulates cargo entry from the ER, while COPI acts more distally, escorting the detached, accelerating tubular entity on its way to joining the Golgi apparatus through microtubule-directed movement.


Subject(s)
COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Microtubules/metabolism , Ubiquitin-Protein Ligases/metabolism , Biological Transport, Active , HeLa Cells , Humans , Protein Transport
8.
Science ; 367(6475)2020 01 17.
Article in English | MEDLINE | ID: mdl-31949053

ABSTRACT

Within cells, the spatial compartmentalization of thousands of distinct proteins serves a multitude of diverse biochemical needs. Correlative super-resolution (SR) fluorescence and electron microscopy (EM) can elucidate protein spatial relationships to global ultrastructure, but has suffered from tradeoffs of structure preservation, fluorescence retention, resolution, and field of view. We developed a platform for three-dimensional cryogenic SR and focused ion beam-milled block-face EM across entire vitreously frozen cells. The approach preserves ultrastructure while enabling independent SR and EM workflow optimization. We discovered unexpected protein-ultrastructure relationships in mammalian cells including intranuclear vesicles containing endoplasmic reticulum-associated proteins, web-like adhesions between cultured neurons, and chromatin domains subclassified on the basis of transcriptional activity. Our findings illustrate the value of a comprehensive multimodal view of ultrastructural variability across whole cells.


Subject(s)
Cells/ultrastructure , Cryoelectron Microscopy/methods , Imaging, Three-Dimensional/methods , Microscopy, Fluorescence/methods , Animals , COS Cells , Cell Adhesion , Cell Line, Tumor , Chlorocebus aethiops , Freezing , HeLa Cells , Humans , Mice
9.
J Cell Biol ; 218(8): 2583-2599, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31227594

ABSTRACT

Lipid droplets (LDs) are neutral lipid storage organelles that transfer lipids to various organelles including peroxisomes. Here, we show that the hereditary spastic paraplegia protein M1 Spastin, a membrane-bound AAA ATPase found on LDs, coordinates fatty acid (FA) trafficking from LDs to peroxisomes through two interrelated mechanisms. First, M1 Spastin forms a tethering complex with peroxisomal ABCD1 to promote LD-peroxisome contact formation. Second, M1 Spastin recruits the membrane-shaping ESCRT-III proteins IST1 and CHMP1B to LDs via its MIT domain to facilitate LD-to-peroxisome FA trafficking, possibly through IST1- and CHMP1B-dependent modifications in LD membrane morphology. Furthermore, LD-to-peroxisome FA trafficking mediated by M1 Spastin is required to relieve LDs of lipid peroxidation. M1 Spastin's dual roles in tethering LDs to peroxisomes and in recruiting ESCRT-III components to LD-peroxisome contact sites for FA trafficking may underlie the pathogenesis of diseases associated with defective FA metabolism in LDs and peroxisomes.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Fatty Acids/metabolism , Lipid Droplets/metabolism , Peroxisomes/metabolism , Spastin/metabolism , ATP Binding Cassette Transporter, Subfamily D, Member 1/metabolism , Adenosine Triphosphatases/metabolism , Amino Acid Motifs , Biological Transport , HeLa Cells , Humans , Hydrolysis , Lauric Acids/metabolism , Models, Biological , Mutant Proteins/metabolism , Oncogene Proteins/metabolism , Spastin/chemistry
10.
Nat Protoc ; 12(5): 916-946, 2017 05.
Article in English | MEDLINE | ID: mdl-28384138

ABSTRACT

Our groups have recently developed related approaches for sample preparation for super-resolution imaging within endogenous cellular environments using correlative light and electron microscopy (CLEM). Four distinct techniques for preparing and acquiring super-resolution CLEM data sets for aldehyde-fixed specimens are provided, including Tokuyasu cryosectioning, whole-cell mount, cell unroofing and platinum replication, and resin embedding and sectioning. The choice of the best protocol for a given application depends on a number of criteria that are discussed in detail. Tokuyasu cryosectioning is relatively rapid but is limited to small, delicate specimens. Whole-cell mount has the simplest sample preparation but is restricted to surface structures. Cell unroofing and platinum replication creates high-contrast, 3D images of the cytoplasmic surface of the plasma membrane but is more challenging than whole-cell mount. Resin embedding permits serial sectioning of large samples but is limited to osmium-resistant probes, and is technically difficult. Expected results from these protocols include super-resolution localization (∼10-50 nm) of fluorescent targets within the context of electron microscopy ultrastructure, which can help address cell biological questions. These protocols can be completed in 2-7 d, are compatible with a number of super-resolution imaging protocols, and are broadly applicable across biology.


Subject(s)
Biological Products/analysis , Cytological Techniques/methods , Microscopy, Electron/methods , Optical Imaging/methods
11.
Angew Chem Int Ed Engl ; 55(5): 1723-7, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26661345

ABSTRACT

The rhodamine system is a flexible framework for building small-molecule fluorescent probes. Changing N-substitution patterns and replacing the xanthene oxygen with a dimethylsilicon moiety can shift the absorption and fluorescence emission maxima of rhodamine dyes to longer wavelengths. Acylation of the rhodamine nitrogen atoms forces the molecule to adopt a nonfluorescent lactone form, providing a convenient method to make fluorogenic compounds. Herein, we take advantage of all of these structural manipulations and describe a novel photoactivatable fluorophore based on a Si-containing analogue of Q-rhodamine. This probe is the first example of a "caged" Si-rhodamine, exhibits higher photon counts compared to established localization microscopy dyes, and is sufficiently red-shifted to allow multicolor imaging. The dye is a useful label for super-resolution imaging and constitutes a new scaffold for far-red fluorogenic molecules.


Subject(s)
Microscopy, Fluorescence/methods , Rhodamines/chemistry , Silicon/analysis
12.
Nat Cell Biol ; 17(7): 880-92, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26053221

ABSTRACT

Focal adhesions (FAs) link the extracellular matrix to the actin cytoskeleton to mediate cell adhesion, migration, mechanosensing and signalling. FAs have conserved nanoscale protein organization, suggesting that the position of proteins within FAs regulates their activity and function. Vinculin binds different FA proteins to mediate distinct cellular functions, but how vinculin's interactions are spatiotemporally organized within FAs is unknown. Using interferometric photoactivation localization super-resolution microscopy to assay vinculin nanoscale localization and a FRET biosensor to assay vinculin conformation, we found that upward repositioning within the FA during FA maturation facilitates vinculin activation and mechanical reinforcement of FAs. Inactive vinculin localizes to the lower integrin signalling layer in FAs by binding to phospho-paxillin. Talin binding activates vinculin and targets active vinculin higher in FAs where vinculin can engage retrograde actin flow. Thus, specific protein interactions are spatially segregated within FAs at the nanoscale to regulate vinculin activation and function.


Subject(s)
Focal Adhesions/metabolism , Nanostructures , Nanotechnology/methods , Vinculin/metabolism , Actins/chemistry , Actins/metabolism , Blotting, Western , Cell Line , Cell Line, Tumor , Fluorescence Resonance Energy Transfer , Focal Adhesions/genetics , Humans , Integrins/chemistry , Integrins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence/methods , Models, Molecular , Mutation , Paxillin/chemistry , Paxillin/genetics , Paxillin/metabolism , Protein Binding , Protein Structure, Tertiary , RNA Interference , Talin/chemistry , Talin/genetics , Talin/metabolism , Vinculin/chemistry , Vinculin/genetics
13.
PLoS Genet ; 11(4): e1005128, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25848771

ABSTRACT

The prokaryotic tubulin homolog, FtsZ, forms a ring-like structure (FtsZ-ring) at midcell. The FtsZ-ring establishes the division plane and enables the assembly of the macromolecular division machinery (divisome). Although many molecular components of the divisome have been identified and their interactions extensively characterized, the spatial organization of these proteins within the divisome is unclear. Consequently, the physical mechanisms that drive divisome assembly, maintenance, and constriction remain elusive. Here we applied single-molecule based superresolution imaging, combined with genetic and biophysical investigations, to reveal the spatial organization of cellular structures formed by four important divisome proteins in E. coli: FtsZ, ZapA, ZapB and MatP. We show that these interacting proteins are arranged into a multi-layered protein network extending from the cell membrane to the chromosome, each with unique structural and dynamic properties. Further, we find that this protein network stabilizes the FtsZ-ring, and unexpectedly, slows down cell constriction, suggesting a new, unrecognized role for this network in bacterial cell division. Our results provide new insight into the structure and function of the divisome, and highlight the importance of coordinated cell constriction and chromosome segregation.


Subject(s)
Bacterial Proteins/metabolism , Cell Division , Cytoskeletal Proteins/metabolism , Escherichia coli/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Escherichia coli/physiology , Escherichia coli/ultrastructure , Escherichia coli Proteins/metabolism , Motion
14.
Nat Methods ; 12(3): 215-8, 4 p following 218, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25581799

ABSTRACT

Fluorescent proteins facilitate a variety of imaging paradigms in live and fixed samples. However, they lose their fluorescence after heavy fixation, hindering applications such as correlative light and electron microscopy (CLEM). Here we report engineered variants of the photoconvertible Eos fluorescent protein that fluoresce and photoconvert normally in heavily fixed (0.5-1% OsO4), plastic resin-embedded samples, enabling correlative super-resolution fluorescence imaging and high-quality electron microscopy.


Subject(s)
Luminescent Proteins/metabolism , Microscopy, Electron, Scanning/methods , Microscopy, Electron, Transmission/methods , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cricetulus , Fluorescence , HeLa Cells , Humans , Luminescent Proteins/genetics , Molecular Imaging/methods , Molecular Sequence Data , Osmium Tetroxide/chemistry , Photochemistry/methods , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
15.
Nat Protoc ; 9(12): 2784-95, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25375992

ABSTRACT

Intracellular recording allows precise measurement and manipulation of individual neurons, but it requires stable mechanical contact between the electrode and the cell membrane, and thus it has remained challenging to perform in behaving animals. Whole-cell recordings in freely moving animals can be obtained by rigidly fixing ('anchoring') the pipette electrode to the head; however, previous anchoring procedures were slow and often caused substantial pipette movement, resulting in loss of the recording or of recording quality. We describe a UV-transparent collar and UV-cured adhesive technique that rapidly (within 15 s) anchors pipettes in place with virtually no movement, thus substantially improving the reliability, yield and quality of freely moving whole-cell recordings. Recordings are first obtained from anesthetized or awake head-fixed rats. UV light cures the thin adhesive layers linking pipette to collar to head. Then, the animals are rapidly and smoothly released for recording during unrestrained behavior. The anesthetized-patched version can be completed in ∼4-7 h (excluding histology) and the awake-patched version requires ∼1-4 h per day for ∼2 weeks. These advances should greatly facilitate studies of neuronal integration and plasticity in identified cells during natural behaviors.


Subject(s)
Cytological Techniques/methods , Electrophysiology/methods , Neurons/physiology , Anesthesia/methods , Animals , Behavior, Animal , Cytological Techniques/instrumentation , Electrodes , Electrophysiology/instrumentation , Equipment Design , Hippocampus/cytology , Motor Activity , Neurons/cytology , Rats, Wistar , Reproducibility of Results , Ultraviolet Rays , Wakefulness
16.
Methods Cell Biol ; 123: 273-94, 2014.
Article in English | MEDLINE | ID: mdl-24974033

ABSTRACT

Many biomolecules in cells can be visualized with high sensitivity and specificity by fluorescence microscopy. However, the resolution of conventional light microscopy is limited by diffraction to ~200-250 nm laterally and >500 nm axially. Here, we describe superresolution methods based on single-molecule localization analysis of photoswitchable fluorophores (PALM: photoactivated localization microscopy) as well as our recent three-dimensional (3D) method (iPALM: interferometric PALM) that allows imaging with a resolution better than 20 nm in all three dimensions. Considerations for their implementations, applications to multicolor imaging, and a recent development that extend the imaging depth of iPALM to ~750 nm are discussed. As the spatial resolution of superresolution fluorescence microscopy converges with that of electron microscopy (EM), direct imaging of the same specimen using both approaches becomes feasible. This could be particularly useful for cross validation of experiments, and thus, we also describe recent methods that were developed for correlative superresolution fluorescence and EM.


Subject(s)
Fluorescent Dyes/chemistry , Single-Cell Analysis/methods , Animals , Cell Line , Fiducial Markers , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Microscopy, Electron , Microscopy, Fluorescence/methods
17.
Nat Methods ; 11(3): 305-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24464288

ABSTRACT

We combine super-resolution localization fluorescence microscopy with transmission electron microscopy of metal replicas to locate proteins on the landscape of the cellular plasma membrane at the nanoscale. We validate robust correlation on the scale of 20 nm by imaging endogenous clathrin (in two and three dimensions) and apply the method to find the previously unknown three-dimensional position of the endocytic protein epsin on clathrin-coated structures at the plasma membrane.


Subject(s)
Gold/chemistry , Membrane Proteins/ultrastructure , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nanotubes/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Adaptor Proteins, Vesicular Transport/ultrastructure , Clathrin/ultrastructure , Humans , Membrane Proteins/metabolism
18.
Science ; 343(6171): 653-6, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24436186

ABSTRACT

The human immunodeficiency virus (HIV) hijacks the endosomal sorting complexes required for transport (ESCRT) to mediate virus release from infected cells. The nanoscale organization of ESCRT machinery necessary for mediating viral abscission is unclear. Here, we applied three-dimensional superresolution microscopy and correlative electron microscopy to delineate the organization of ESCRT components at HIV assembly sites. We observed ESCRT subunits localized within the head of budding virions and released particles, with head-localized levels of CHMP2A decreasing relative to Tsg101 and CHMP4B upon virus abscission. Thus, the driving force for HIV release may derive from initial scaffolding of ESCRT subunits within the viral bud interior followed by plasma membrane association and selective remodeling of ESCRT subunits.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , HIV Infections/virology , HIV-1/physiology , Virion/physiology , Virus Assembly , HIV-1/metabolism , Humans , Imaging, Three-Dimensional/methods , Microscopy/methods , Protein Subunits/metabolism , Virion/metabolism , Virus Release , gag Gene Products, Human Immunodeficiency Virus/metabolism
19.
PLoS One ; 8(10): e77209, 2013.
Article in English | MEDLINE | ID: mdl-24204771

ABSTRACT

The ability to localize proteins precisely within subcellular space is crucial to understanding the functioning of biological systems. Recently, we described a protocol that correlates a precise map of fluorescent fusion proteins localized using three-dimensional super-resolution optical microscopy with the fine ultrastructural context of three-dimensional electron micrographs. While it achieved the difficult simultaneous objectives of high photoactivated fluorophore preservation and ultrastructure preservation, it required a super-resolution optical and specialized electron microscope that is not available to many researchers. We present here a faster and more practical protocol with the advantage of a simpler two-dimensional optical (Photoactivated Localization Microscopy (PALM)) and scanning electron microscope (SEM) system that retains the often mutually exclusive attributes of fluorophore preservation and ultrastructure preservation. As before, cryosections were prepared using the Tokuyasu protocol, but the staining protocol was modified to be amenable for use in a standard SEM without the need for focused ion beam ablation. We show the versatility of this technique by labeling different cellular compartments and structures including mitochondrial nucleoids, peroxisomes, and the nuclear lamina. We also demonstrate simultaneous two-color PALM imaging with correlated electron micrographs. Lastly, this technique can be used with small-molecule dyes as demonstrated with actin labeling using phalloidin conjugated to a caged dye. By retaining the dense protein labeling expected for super-resolution microscopy combined with ultrastructural preservation, simplifying the tools required for correlative microscopy, and expanding the number of useful labels we expect this method to be accessible and valuable to a wide variety of researchers.


Subject(s)
Actins/ultrastructure , Microscopy, Electron, Scanning/instrumentation , Microscopy, Fluorescence/instrumentation , Mitochondria/ultrastructure , Nuclear Lamina/ultrastructure , Peroxisomes/ultrastructure , Animals , Cryoultramicrotomy , Imaging, Three-Dimensional , Mice , Microscopy, Electron, Scanning/methods , Microscopy, Fluorescence/methods , Microtomy/methods , NIH 3T3 Cells , Phalloidine/chemistry , Staining and Labeling/methods
20.
Nat Commun ; 3: 1154, 2012.
Article in English | MEDLINE | ID: mdl-23093191

ABSTRACT

The molecular mechanism responsible for capturing, sorting and retrieving vesicle membrane proteins following triggered exocytosis is not understood. Here we image the post-fusion release and then capture of a vesicle membrane protein, the vesicular acetylcholine transporter, from single vesicles in living neuroendocrine cells. We combine these measurements with super-resolution interferometric photo-activation localization microscopy and electron microscopy, and modelling to map the nanometer-scale topography and architecture of the structures responsible for the transporter's capture following exocytosis. We show that after exocytosis, the transporter rapidly diffuses into the plasma membrane, but most travels only a short distance before it is locally captured over a dense network of membrane-resident clathrin-coated structures. We propose that the extreme density of these structures acts as a short-range diffusion trap. They quickly sequester diffusing vesicle material and limit its spread across the membrane. This system could provide a means for clathrin-mediated endocytosis to quickly recycle vesicle proteins in highly excitable cells.


Subject(s)
Membrane Fusion/physiology , Membrane Proteins/physiology , Vesicular Acetylcholine Transport Proteins/physiology , Animals , Cell Membrane/physiology , Cell Membrane/ultrastructure , Clathrin/physiology , Clathrin/ultrastructure , Endocytosis/physiology , Exocytosis/physiology , Membrane Proteins/ultrastructure , Microscopy, Electron , Microscopy, Interference/methods , PC12 Cells/physiology , Rats , Synaptic Vesicles/physiology , Synaptic Vesicles/ultrastructure , Vesicular Acetylcholine Transport Proteins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...