Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biomed Opt Express ; 14(1): 429-440, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36698655

ABSTRACT

We describe a dedicated microscope for automated sequential localization microscopy which we term Sequential Super-resolution Microscope (SeqSRM). This microscope automates precise stage stabilization on the order of 5-10 nanometers and data acquisition of all user-selected cells on a coverslip, limiting user interaction to only cell selection and buffer exchanges during sequential relabeling. We additionally demonstrate that nanometer-scale changes to cell morphology affect the fidelity of the resulting multi-target super-resolution overlay reconstructions generated by sequential super-resolution microscopy, and that regions affected by these shifts can be reliably detected and masked out using brightfield images collected periodically throughout the experiment. The SeqSRM enables automated multi-target imaging on multiple user-selected cells without the need for multiple distinct fluorophores and emission channels, while ensuring that the resulting multi-target localization data accurately reflect the relative organization of the underlying targets.

4.
PLoS One ; 16(1): e0246138, 2021.
Article in English | MEDLINE | ID: mdl-33508018

ABSTRACT

Visualizing actin filaments in fixed cells is of great interest for a variety of topics in cell biology such as cell division, cell movement, and cell signaling. We investigated the possibility of replacing phalloidin, the standard reagent for super-resolution imaging of F-actin in fixed cells, with the actin binding peptide 'lifeact'. We compared the labels for use in single molecule based super-resolution microscopy, where AlexaFluor 647 labeled phalloidin was used in a dSTORM modality and Atto 655 labeled lifeact was used in a single molecule imaging, reversible binding modality. We found that imaging with lifeact had a comparable resolution in reconstructed images and provided several advantages over phalloidin including lower costs, the ability to image multiple regions of interest on a coverslip without degradation, simplified sequential super-resolution imaging, and more continuous labeling of thin filaments.


Subject(s)
Actin Cytoskeleton/pathology , Carbocyanines/chemistry , Phalloidine/chemistry , Actin Cytoskeleton/chemistry , HeLa Cells , Humans , Microscopy, Fluorescence
5.
Methods Mol Biol ; 2038: 131-150, 2019.
Article in English | MEDLINE | ID: mdl-31407282

ABSTRACT

Single-molecule resolution imaging has become an important tool in the study of cell biology. Aptamer-based approaches (e.g., MS2 and PP7) allow for detection of single RNA molecules in living cells and have been used to study various aspects of mRNA metabolism, including mRNP nuclear export. Here we outline an imaging protocol for the study of interactions between mRNPs and nuclear pore complexes (NPCs) in the yeast S. cerevisiae, including mRNP export. We describe in detail the steps that allow for high-resolution live-cell mRNP imaging and measurement of mRNP interactions with NPCs using simultaneous two-color imaging. Our protocol discusses yeast strain construction, choice of marker proteins to label the nuclear pore complex, as well as imaging conditions that allow high signal-to-noise data acquisition. Moreover, we describe various aspects of postacquisition image analysis for single molecule tracking and image registration allowing for the characterization of mRNP-NPC interactions.


Subject(s)
Microscopy, Fluorescence , Molecular Imaging/methods , Nuclear Pore/metabolism , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Single Molecule Imaging/methods , Active Transport, Cell Nucleus , Gene Expression Regulation, Fungal , Nuclear Pore/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Time Factors
6.
Dev Cell ; 49(1): 130-144.e6, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30827897

ABSTRACT

Syntaxin 17 (Stx17) has been implicated in autophagosome-lysosome fusion. Here, we report that Stx17 functions in assembly of protein complexes during autophagy initiation. Stx17 is phosphorylated by TBK1 whereby phospho-Stx17 controls the formation of the ATG13+FIP200+ mammalian pre-autophagosomal structure (mPAS) in response to induction of autophagy. TBK1 phosphorylates Stx17 at S202. During autophagy induction, Stx17pS202 transfers from the Golgi, where its steady-state pools localize, to the ATG13+FIP200+ mPAS. Stx17pS202 was in complexes with ATG13 and FIP200, whereas its non-phosphorylatable mutant Stx17S202A was not. Stx17 or TBK1 knockouts blocked ATG13 and FIP200 puncta formation. Stx17 or TBK1 knockouts reduced the formation of ATG13 protein complexes with FIP200 and ULK1. Endogenous Stx17pS202 colocalized with LC3B following induction of autophagy. Stx17 knockout diminished LC3 response and reduced sequestration of the prototypical bulk autophagy cargo lactate dehydrogenase. We conclude that Stx17 is a TBK1 substrate and that together they orchestrate assembly of mPAS.


Subject(s)
Autophagy/genetics , Multiprotein Complexes/genetics , Protein Serine-Threonine Kinases/genetics , Qa-SNARE Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Autophagosomes/metabolism , Autophagy-Related Proteins/genetics , Gene Knockout Techniques , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Humans , Lysosomes/metabolism , Membrane Fusion/genetics , Multiprotein Complexes/metabolism , Mutation/genetics , Phosphorylation , Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics
7.
PLoS One ; 13(8): e0203291, 2018.
Article in English | MEDLINE | ID: mdl-30169528

ABSTRACT

Sequential labeling and imaging in fluorescence microscopy allows the imaging of multiple structures in the same cell using a single fluorophore species. In super-resolution applications, the optimal dye suited to the method can be chosen, the optical setup can be simpler and there are no chromatic aberrations between images of different structures. We describe a method based on DNA strand displacement that can be used to quickly and easily perform the labeling and removal of the fluorophores during each sequence. Site-specific tags are conjugated with unique and orthogonal single stranded DNA. Labeling for a particular structure is achieved by hybridization of antibody-bound DNA with a complimentary dye-labeled strand. After imaging, the dye is removed using toehold-mediated strand displacement, in which an invader strand competes off the dye-labeled strand than can be subsequently washed away. Labeling and removal of each DNA-species requires only a few minutes. We demonstrate the concept using sequential dSTORM super-resolution for multiplex imaging of subcellular structures.


Subject(s)
DNA, Single-Stranded , Microscopy, Fluorescence/methods , Antibodies , Clathrin , Click Chemistry , DNA, Single-Stranded/chemistry , Fluorescent Dyes , HeLa Cells , Humans , Intracellular Space , Motion , Tissue Fixation , Tubulin
8.
J Cell Biol ; 217(3): 997-1013, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29420192

ABSTRACT

Autophagy is a conserved eukaryotic process with metabolic, immune, and general homeostatic functions in mammalian cells. Mammalian autophagosomes fuse with lysosomes in a SNARE-driven process that includes syntaxin 17 (Stx17). How Stx17 translocates to autophagosomes is unknown. In this study, we show that the mechanism of Stx17 recruitment to autophagosomes in human cells entails the small guanosine triphosphatase IRGM. Stx17 directly interacts with IRGM, and efficient Stx17 recruitment to autophagosomes requires IRGM. Both IRGM and Stx17 directly interact with mammalian Atg8 proteins, thus being guided to autophagosomes. We also show that Stx17 is significant in defense against infectious agents and that Stx17-IRGM interaction is targeted by an HIV virulence factor Nef.


Subject(s)
Autophagosomes/metabolism , Autophagy-Related Protein 8 Family/metabolism , GTP-Binding Proteins/metabolism , Qa-SNARE Proteins/metabolism , Autophagy-Related Protein 8 Family/genetics , GTP-Binding Proteins/genetics , HEK293 Cells , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/genetics , HIV-1/metabolism , HeLa Cells , Humans , Protein Transport/genetics , Qa-SNARE Proteins/genetics , THP-1 Cells , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/metabolism
9.
Autophagy ; 13(6): 1084-1085, 2017 Jun 03.
Article in English | MEDLINE | ID: mdl-28368721

ABSTRACT

Macroautophagy/autophagy plays a role in unconventional secretion of leaderless cytosolic proteins. Whether and how secretory autophagy diverges from conventional degradative autophagy is unclear. We have shown that the prototypical secretory autophagy cargo IL1B/IL-1ß (interleukin 1 ß) is recognized by TRIM16, and that this first to be identified secretory autophagy receptor interacts with the R-SNARE SEC22B to jointly deliver cargo to the MAP1LC3B-II-positive sequestration membranes. Cargo secretion is unaffected by knockdowns of STX17, a SNARE catalyzing autophagosome-lysosome fusion as a prelude to cargo degradation. Instead, SEC22B in combination with plasma membrane syntaxins completes cargo secretion. Thus, secretory autophagy diverges from degradative autophagy by using specialized receptors and a dedicated SNARE machinery to bypass fusion with lysosomes.


Subject(s)
Autophagy , Secretory Pathway , Humans , Lysosomes/metabolism , Membrane Fusion , Models, Biological , Phagosomes/metabolism , SNARE Proteins/metabolism
10.
EMBO J ; 36(1): 42-60, 2017 01 04.
Article in English | MEDLINE | ID: mdl-27932448

ABSTRACT

Autophagy is a process delivering cytoplasmic components to lysosomes for degradation. Autophagy may, however, play a role in unconventional secretion of leaderless cytosolic proteins. How secretory autophagy diverges from degradative autophagy remains unclear. Here we show that in response to lysosomal damage, the prototypical cytosolic secretory autophagy cargo IL-1ß is recognized by specialized secretory autophagy cargo receptor TRIM16 and that this receptor interacts with the R-SNARE Sec22b to recruit cargo to the LC3-II+ sequestration membranes. Cargo secretion is unaffected by downregulation of syntaxin 17, a SNARE promoting autophagosome-lysosome fusion and cargo degradation. Instead, Sec22b in combination with plasma membrane syntaxin 3 and syntaxin 4 as well as SNAP-23 and SNAP-29 completes cargo secretion. Thus, secretory autophagy utilizes a specialized cytosolic cargo receptor and a dedicated SNARE system. Other unconventionally secreted cargo, such as ferritin, is secreted via the same pathway.


Subject(s)
Autophagy , DNA-Binding Proteins/metabolism , Interleukin-1beta/metabolism , Microtubule-Associated Proteins/metabolism , R-SNARE Proteins/metabolism , Transcription Factors/metabolism , Cell Line , Ferritins/metabolism , Humans , Monocytes/metabolism , Qa-SNARE Proteins/metabolism , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...