Abstract
The actin cytoskeleton plays multiple critical roles in cells, from cell migration to organelle dynamics. The small and transient actin structures regulating organelle dynamics are difficult to detect with fluorescence microscopy, and the limited resolution of fluorescence microscopy makes it difficult to determine whether actin filaments are directly associated with specific membranes. To address these limitations, we developed an approach using fluorescent protein-tagged actin nanobodies targeted to organelle membranes to enable live cell imaging of sub-organellar actin dynamics with unprecedented spatiotemporal resolution.
Introduction
The critical role of the actin cytoskeleton in organelle dynamics is largely accepted, but poorly understood. The precise spatiotemporal dynamics of actin at organelle membranes remain particularly unclear due to the combined limitations of currently available actin probes and imaging approaches. For fluorescence microscopy approaches, imaging smaller actin structures in the cell suffers from an enormous background signal issue - the high signal from the dense meshwork of actin filaments throughout the cell overwhelms the signal from the relatively small, transient actin structures associated with organelle dynamics. Furthermore, the limitations in resolution make it difficult to conclude whether any actin filaments are directly associated with the organelle. Here we employ fluorescent protein-tagged actin nanobodies, aka “actin chromobodies” (AC)1,2, fused to organelle membrane targeting sequences to facilitate live cell imaging of sub-organellar actin dynamics with high spatiotemporal resolution. Using these probes, we imaged the spatiotemporal dynamics of mitochondria- and endoplasmic reticulum (ER)-associated actin accumulation at mitochondrial fission sites.
Results
We hypothesized that AC probes with organelle membrane targeting sequences could be used to visualize actin filaments closely associated with the target organelle membrane. Given the high mobility of membrane-anchored proteins in a lipid bilayer, an F-actin binding probe containing only a membrane-anchoring sequence and a fluorescent protein tag should quickly accumulate at sites of F-actin enrichment near the membrane by virtue of its actin-binding activity (Fig. 1A). To test this hypothesis, we generated constructs containing the yeast Fis1 mitochondrial outer membrane or Cytb5ER endoplasmic reticulum (ER) minimal C-terminus tail membrane targeting sequences fused to the cytoplasm-facing actin nanobody and tagGFP (“AC-mito” and “AC-ER”)3. Live cell Airyscan confocal imaging of cells expressing AC-mito or AC-ER counterstained with MitoTracker dye revealed specific regions of AC-mito accumulation on the surfaces of mitochondria, and similarly for AC-ER on the ER (Fig. 1B and Supp. Videos 1–4). To rule out the possibility the membrane targeting sequences we used were causing the probe to accumulate in specific regions independent of actin-binding activity, we co-transfected AC-mito or AC-ER with control probes: mCherry-tagged mitochondrial (Fis1) and ER (Cytb5ER) membrane targeting sequences (mCherry-mito and mCherry-ER). As expected, the mCherry-mito and mCherry-ER signals were evenly distributed along their respective organelle membranes, with no obvious accumulation in any specific regions. In contrast, the co-expressed AC-mito and AC-ER constructs displayed significant accumulation in specific regions on their respective organelles (Fig. 1B), as quantified by the coefficient of variance (see Methods for more details). Cells expressing AC-ER counterstained with MitoTracker revealed high AC-ER accumulation at ER-mitochondria contacts compared to the mCherry-ER control probe (Fig. 1B, Extended Data Fig. 1, and Supp. Videos 3–4). Overall, AC probe labeling appears to provide both more specificity and sensitivity than phalloidin staining for identifying organelle-associated actin (Extended Data Fig. 2). Importantly, there was no detected increase in actin on organelles in cells expressing the AC probes (Extended Data Fig. 2C). Similarly, we found no change in endogenous Fis1 localization to mitochondria in AC-mito expressing cells (Extended Data Fig. 3). Using a different membrane targeting sequence (Cytb5mito3) and/or a different F-actin probe (LifeAct4) yielded similar results (Extended Data Fig. 4).
Fluorescence recovery after photobleaching (FRAP) experiments showed that both the AC and mCherry control probes are highly mobile on the membrane, but only the AC probes exhibit lower mobility at sites of AC specific accumulation (Fig. 1C, Extended Data Fig. 5, Supp. Videos 5, 6). However, the AC probe mobility was similar to the control probes in diffuse regions lacking AC accumulation (Extended Data Fig. 5), consistent with the model in which AC accumulation is indicative of immobilization by F-actin binding activity (Fig. 1A)5,6. To further test whether sub-organellar AC accumulation is dependent on F-actin, we treated AC expressing cells with the F-actin depolymerizing drug Latrunculin B (LatB). LatB treatment significantly altered not only the distribution but also increased the mobility of the AC probes (Extended Data Fig. 6). Taken together, these results strongly support the conclusion that AC probe accumulation and mobility is dependent on F-actin, yet the AC binding dynamics are still relatively fast compared to actin filament turnover rates7–9.
Given their high mobility, we hypothesized AC probes should reveal dynamic changes in F-actin (dis)assembly on their respective organelles. In HeLa cells, F-actin “waves” cycle around the cell, decorating subpopulations of mitochondria prior to their fission and subsequent fusion10. As expected, we found coaccumulation of F-actin and the AC probes (but not the control probes) on specific regions of the organelles (Fig. 2). Timelapse imaging of cells labeled with LifeAct, the AC probes, and a mitochondrial matrix marker revealed fluctuations in actin accumulation on mitochondrial subpopulations as actin waves cycled throughout the cell (Fig. 2B, Extended Data Fig. 7, Supp. Videos 7–9). Fluorescence intensity changes in AC probe levels matched those of LifeAct, and mitochondrial fission occurred after actin accumulation, as previously reported (Fig. 2D, Supp. Video 10). The wave cycling rates matched those of cells not expressing AC probes, and neighboring cells expressing varying levels of the AC probe also had matching rates. Increased AC accumulation around fragmented mitochondria induced by ionomycin treatment was also evident. There was no difference in the rate of ionomycin-induced mitochondrial fragmentation in cells expressing AC probes compared to neighboring untransfected cells (Extended Data. Fig 8, Supp. Videos 11, 12). Overall, these results show the AC probes reliably label dynamic actin (dis)assembly without altering the kinetics or mitochondrial behavior in normal or stressed conditions.
Previous studies demonstrated a role for actin in driving mitochondrial fission at ER-mitochondria contact sites, likely with actin playing a role in force generation and Drp1 recruitment and activation11–18. To determine the kinetics of mitochondria- and ER-associated actin accumulation at mitochondrial fission sites, we performed live imaging of cells expressing AC-mito and AC-ER counterstained with MitoTracker. Mitochondrial fission sites displayed accumulation of both AC-mito and AC-ER prior to Drp1- and ER-mediated fission. (Fig. 3, Extended Data Figs. 9, 10, Supp Figs. 1–8, Supp. Videos 13–19). Approximately 5% of cells transfected with AC-mito expressed off a CMV promoter displayed overexpression artifacts (i.e. increased clustering and reduced mobility of mitochondria) (Supp. Video 20). To address this, we expressed AC-mito using a ubiquitin UbC promoter. This dramatically reduced the prevalence of overexpression artifacts (less than 0.01% of transfected cells). We did not detect a change in mitochondrial membrane potential, fission rate, or ER-mitochondria contacts in cells expressing the AC probes (Extended Data Fig. 9). In cells expressing both AC-mito and AC-ER, we invariably observed AC-mito accumulation prior to AC-ER (Fig. 3D, Supp. Fig. 9, Supp. Video 21).
Conclusion
Membrane-anchored AC probes provide a novel tool for studying membrane-associated actin dynamics with much higher effective resolution than cytoplasmic actin probes. In addition to the plasma membrane19, the variety of subcellular compartments, protein targets, and corresponding subcellular or sub-organellar processes that can be studied will increase as additional nanobodies or other kinds of protein binding motifs and subcellular membrane targeting sequences are developed. In addition, future studies switching the fluorescent markers for EM compatible probes, and/or proximity markers for downstream mass spectrometry analyses such as APEX220 or TurboID21 will likely reveal new insights into fundamental processes and molecular mechanisms in actin and organelle cell biology.
Methods
Cell culture.
U2OS, HeLa, and Hap1 cells were purchased from ATCC. HeLa cells stably expressing LifeAct-mCherry were a gift from the Wedlich-Soldner lab22. Cells were grown in DMEM supplemented with 10% fetal bovine serum at 37°C with 5% CO2. Cells were transfected with Lipofectamine 2000 (ThermoFisher). Cells were plated onto either 8-well #1.5 imaging chambers or #1.5 35mm dishes (Cellvis) that were coated with 10µg/mL fibronectin in PBS at 37°C for 30 minutes prior to plating. 50nM MitoTracker Deep Red (ThermoFisher) was added for 30 minutes then washed for at least 30 minutes to allow for recovery time before imaging in FluoroBrite (ThermoFisher) medium.
Airyscan confocal imaging.
Cells were imaged with a Plan-Apochromat 63x/1.4NA oil objective on an inverted Zeiss 880 LSM Airyscan confocal microscope with the environmental control system supplying 37°C, 5% CO2 and humidity for live cell imaging. The GFP channels were imaged with a 488nm laser line at ~500nW laser power. The mCherry or tagRFP channels were imaged with 561nm laser at ~1µW laser power. The MitoTracker Deep Red channel was imaged with ~250nW laser power. For timelapse imaging, the zoom factor was set between 3x-6x to increase the frame rate. In all cases, the maximum pixel dwell time (~0.684µs/pixel) and 2x Nyquist optimal pixel size (~40nm/pixel) was used.
Spinning disk confocal imaging.
Cells were imaged with a Plan-Apochromat 40x/1.3NA oil objective on a Zeiss CSU Spinning Disk Confocal Microscope with a CSU-X1 Yokogawa spinning disk scan head on a Prime 95B sCMOS camera (Teledyne Photometrics). The 488/561/647nm laser powers were set at 100/200/20µW with 300/300/150ms exposure times, respectively.
Antibodies.
We used the rabbit anti-Fis1 antibody against the N-terminal cytoplasmic facing side of the human Fis1 protein, made by Prestige Antibodies Powered by Atlas Antibodies (Sigma-Aldrich, catalog #: HPA017430). The amino acid sequence of the antigen is: MEAVLNELVSVEDLLKFEKKFQSEKAAGSVSKSTQFEYAWCLVRSKYNDDIRKGIVLLEELLPKGS KEEQRDYVFYLAVGNYRLKEYEKALKYVRGLLQTEPQNNQAKELERLIDKAMKKD.
Immunofluorescence.
Cells were washed in PBS then fixed with 4% PFA for 30 minutes before permeabilization with 0.1% Triton X-100 for 30 minutes. Cells were then blocked overnight with 4% BSA at 4°C. Cells were then incubated with primary antibody for 2 hours, rinsed 3x with PBS for 10 minutes each, then incubated with secondary antibodies (Jackson Immunoresearch Laboratories) for 1 hour, rinsed 3x with PBS for 10 minutes each, then counterstained with Alexa405-phalloidin (ThermoFisher) for 30 minutes, then rinsed with PBS 3x for 10 minutes each, then mounted with ProLong Glass antifade reagent (ThermoFisher).
Image processing and analysis.
After acquisition, images were Airyscan processed using the auto-filter 2D-SR settings in Zen Blue (ZEISS). All images were post-processed and analyzed using Imaris (BITPLANE) and Fiji software23. All images shown are from single focal planes unless stated otherwise.
Data quantification and statistics.
All line scans were normalized and plotted in Excel. All statistical analyses and graphs were generated using GraphPad Prism 8 software. All graphs display horizontal lines marking average values and error bars indicating standard deviation.
AC probe “accumulation” calculation (Supp. Fig. 5):
Using Fiji, a square selection was drawn around a region with obvious AC probe accumulation (1.5 µm2 for mitochondria, 0.6 µm2 for ER). The mean pixel intensity of the AC probe and mCherry control probe within the selection was measured. Another square of equal dimensions was drawn in an adjacent area with mCherry signal but without obvious AC probe accumulation. Mean pixel intensity was also measured in this region. The mean pixel intensity in the accumulated region was then divided by the mean pixel intensity in the region without accumulation.
Determination of coefficient of variance:
We measured the extent of sub-organellar “accumulation” by quantifying the coefficient of variance as follows: In Fiji, the mCherry-mito or mCherry-ER signal was used to generate a mask of the mitochondria or ER, respectively. A selection was generated based on this mask (see Fig. 2d and Supp. Fig. 14a). The mean pixel intensity and standard deviation within the mask were measured. Coefficient of variance was determined by dividing the standard deviation by the mean pixel intensity.
Determination of % overlap:
Masks of ER, mitochondria, actin, and AC/mCherry probes were generated in Fiji. For AC probes, thresholding was set to mask only the top 25% of AC probe signal based on maximum pixel intensity (see the “clipped” panels in Supp. Fig. 2). All other masks were generated using default thresholding settings in Fiji, which uses the IsoData algorithm developed by Ridler and Calvard24. The integrated density of each mask was calculated. Areas of overlap between masks were generated using Fiji’s “Image Calculator” and the integrated density of these areas was also measured. These values were used to calculate the % overlap (i.e. integrated density for area of overlap between AC-mito and mitochondria divided by integrated density for mitochondria area yields % of mitochondria overlapped by AC-mito). This percentage was interpreted as the probability of AC probes localizing to fission sites by chance (Fig. 3c).
Colocalization analysis:
Pearson’s correlation coefficient was determined using Fiji’s “Coloc 2” plugin (https://imagej.net/Coloc_2). For analysis of colocalization in regions with or without AC probe accumulation (Fig. 2c), a square selection was drawn around a region with obvious AC probe accumulation and an equal-sized square was drawn in an adjacent region without AC probe accumulation. Square sizes were 1.5 µm2 for AC-mito analysis and 0.6 µm2 for AC-ER analysis. For mitochondria-ER contact analysis (Supp. Fig. 11c), a peripheral region (98.5 µm2) of the cell containing mitochondria and ER was selected. This was done to prevent artificially high values resulting from the large amount of ER and mitochondria overlap typically observed in the perinuclear region, which is too dense to resolve by Airyscan confocal microscopy.
FRAP analysis:
FRAP experiments were performed on a Zeiss 880 Airyscan confocal microscope using a Plan-Apochromat 63x/1.4 Oil DIC objective. Transfected cells were maintained at 37°C with 5% CO2 in FluoroBrite DMEM (Gibco) and 10% FBS (VWR) culture medium. 488nm and 561nm excitation laser lines and Airyscan detectors were driven by Zeiss Zen black software. FRAP experiments were done in one focal plane, using the following conditions: 3 pre-bleach frames were acquired at maximum speed with 488nm laser at 25µW and 561nm laser at 13µW power. The photobleaching of selected regions was done with 488nm laser at 5mW power at maximum speed for 30 iterations. The post-bleach acquisition was done with 488nm at pre-bleach imaging settings for 100 frames. The fluorescence intensity of the acquired images were quantified in Fiji following the principles as outlined in Lippincott-Schwartz et al. 201825. The mobile fraction was determined as the percentage of fluorescence recovery at full recovery. The t1/2 was determined as the time taken for the fluorescence intensity in the bleached region to recover to 50% of the full recovery value after bleaching. If objects moved in or out of the ROI during the recovery phase, they were not included in our analyses.
Actin wave pixel intensity measurements:
Square selections were made in Fiji (3 selections per cell) and the mean pixel intensity was measured over time. The selections used were 10.1 µm2 for data collected using Airyscan confocal and 32.8 µm2 for data collected using spinning disk confocal microscopy.
Mitochondrial membrane potential:
The mean threshold value for each cell was obtained through thresholding to include mitochondria using the MitoTracker Deep Red channel. The average pixel intensity in the thresholded region was measured. The relative intensity for each cell was then calculated by dividing each cell’s mean threshold value by the average mean threshold value of all un-transfected cells within the same image. The relative signal intensity of each transfected and un-transfected cell was then plotted and graphed.
Plasmids.
Drp1-mCherry was a kind gift from Gia Voeltz (Addgene plasmid #49152). mCherry-Cytob5RR was a gift from Nica Borgese26. Halo-Sec6127, Halo-Fis1 (Addgene plasmid #111136), LifeAct-mScarlet (Addgene plasmid #85054), and mTagBFP2-mito28 used for experiments shown in Figure 2 were gifts from the Lippincott-Schwartz lab. All custom actin nanobody probes were generated starting from the commercial vector of actin chromobody-tagGFP or actin chromobody-tagRFP (ChromoTek) and cloned via the BglII and NotI restriction sites. The following amino acid sequences were attached to the C-terminal of the actin chromobody probes to target the protein either to mitochondria or the ER:
Fis1 (AC-mito and LifeAct-GFP-Fis1):
IQKETLKGVVVAGGVLAGAVAVASFFLRNKRR3
Cytb5mito (aka “Cytob5RR”) (AC-GFP-Cytb5mito and LifeAct-GFP-Cytb5mito):
FEPSETLITTVESNSSWWTNWVIPAISALVVALMYRR29
Cytb5ER (AC-ER):
IDSSSSWWTNWVIPAISAVAVALMYRLYMAED3
LifeAct-GFP-Fis1, LifeAct-GFP-Cytb5mito, and AC-GFP-Cytb5mito were generated using PFU Ultra II for megaprimer PCR insertion30. The PCR primers, intended modifications, insert templates, and destination plasmids are listed in Supplementary Table 1. All constructs were sequenced completely across their coding region.
Extended Data
Supplementary Material
Acknowledgements
We are grateful to Stephanie Harada (Salk Institute for Biological Studies) for help with the cartoon diagram for Fig. 1. We would like to also thank Christopher Obara and Jennifer Lippincott-Schwartz (Janelia Farms) for critical feedback and suggestions on the manuscript. We would also like to thank our reviewers for critical comments that greatly improved the quality, rigor, and interpretability of our work. This work was also greatly improved by feedback on our initial bioRxiv preprint received via Twitter. The Waitt Advanced Biophotonics Center is funded by the Waitt Foundation and Core Grant application NCI CCSG (CA014195). This work was supported by the Transgenic Core Facility of the Salk Institute with funding from NIH-NCI CCSG: P30 014195. R.G. laboratory is funded by grants from HFSP RGP0021/2016 and the Cluster of Excellence CIBSS- Centre for Integrative Biological Signaling Studies (DFG, EXC-2189). G.S.S. is supported by NIH grant no. R01 AR069876 and the Salk Institute Audrey Geisel Chair in Biomedical Science. O.A.Q. lab is supported by NIGMS Grant R15 GM119077 and by funding from the University of Richmond School of Arts & Sciences.
Footnotes
Ethics Declaration
The authors declare no competing interests.
Data Availability
The original source data for quantification and the raw imaging data used for all the presented figures and videos will be made available from zenodo.org doi: 10.5281/zenodo.2851619.
References
- 1.Rocchetti A, Hawes C & Kriechbaumer V Fluorescent labelling of the actin cytoskeleton in plants using a cameloid antibody. Plant Methods 10, 12, doi: 10.1186/1746-4811-10-12 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Melak M, Plessner M & Grosse R Actin visualization at a glance. J Cell Sci 130, 525–530, doi: 10.1242/jcs.189068 (2017). [DOI] [PubMed] [Google Scholar]
- 3.Rapaport D Finding the right organelle. Targeting signals in mitochondrial outer-membrane proteins. EMBO reports 4, 948–952, doi: 10.1038/sj.embor.embor937 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Riedl J et al. Lifeact: a versatile marker to visualize F-actin. Nat Methods 5, 605–607, doi: 10.1038/nmeth.1220 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sprague BL & McNally JG FRAP analysis of binding: proper and fitting. Trends in cell biology 15, 84–91, doi: 10.1016/j.tcb.2004.12.001 (2005). [DOI] [PubMed] [Google Scholar]
- 6.Sprague BL, Pego RL, Stavreva DA & McNally JG Analysis of binding reactions by fluorescence recovery after photobleaching. Biophys J 86, 3473–3495, doi: 10.1529/biophysj.103.026765 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Panza P, Maier J, Schmees C, Rothbauer U & Sollner C Live imaging of endogenous protein dynamics in zebrafish using chromobodies. Development 142, 1879–1884, doi: 10.1242/dev.118943 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Shirai YM et al. Cortical actin nodes: Their dynamics and recruitment of podosomal proteins as revealed by super-resolution and single-molecule microscopy. PloS one 12, e0188778, doi: 10.1371/journal.pone.0188778 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bisaria A, Hayer A, Garbett D, Cohen D & Meyer T Membrane-proximal F-actin restricts local membrane protrusions and directs cell migration. Science 368, 1205, doi: 10.1126/science.aay7794 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Moore AS, Wong YC, Simpson CL & Holzbaur EL Dynamic actin cycling through mitochondrial subpopulations locally regulates the fission-fusion balance within mitochondrial networks. Nature communications 7, 12886, doi: 10.1038/ncomms12886 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Manor U et al. A mitochondria-anchored isoform of the actin-nucleating spire protein regulates mitochondrial division. eLife 4, doi: 10.7554/eLife.08828 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chakrabarti R et al. INF2-mediated actin polymerization at the ER stimulates mitochondrial calcium uptake, inner membrane constriction, and division. J Cell Biol 217, 251–268, doi: 10.1083/jcb.201709111 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Korobova F, Ramabhadran V & Higgs HN An actin-dependent step in mitochondrial fission mediated by the ER-associated formin INF2. Science 339, 464–467, doi: 10.1126/science.1228360 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Korobova F, Gauvin TJ & Higgs HN A role for myosin II in mammalian mitochondrial fission. Curr Biol 24, 409–414, doi: 10.1016/j.cub.2013.12.032 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yang C & Svitkina TM Ultrastructure and dynamics of the actin-myosin II cytoskeleton during mitochondrial fission. Nat Cell Biol 21, 603–613, doi: 10.1038/s41556-019-0313-6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.De Vos KJ, Allan VJ, Grierson AJ & Sheetz MP Mitochondrial function and actin regulate dynamin-related protein 1-dependent mitochondrial fission. Curr Biol 15, 678–683, doi: 10.1016/j.cub.2005.02.064 (2005). [DOI] [PubMed] [Google Scholar]
- 17.Li S et al. Transient assembly of F-actin on the outer mitochondrial membrane contributes to mitochondrial fission. J Cell Biol 208, 109–123, doi: 10.1083/jcb.201404050 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ji WK, Hatch AL, Merrill RA, Strack S & Higgs HN Actin filaments target the oligomeric maturation of the dynamin GTPase Drp1 to mitochondrial fission sites. eLife 4, e11553, doi: 10.7554/eLife.11553 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bisaria A, Hayer A, Garbett D, Cohen D & Meyer T Membrane proximal F-actin restricts local membrane protrusions and directs cell migration. BioRxiv, 705509 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lam SS et al. Directed evolution of APEX2 for electron microscopy and proximity labeling. Nat Methods 12, 51–54, doi: 10.1038/nmeth.3179 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Branon TC et al. Efficient proximity labeling in living cells and organisms with TurboID. Nature biotechnology 36, 880–887, doi: 10.1038/nbt.4201 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wales P et al. Calcium-mediated actin reset (CaAR) mediates acute cell adaptations. eLife 5, doi: 10.7554/eLife.19850 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Schindelin J et al. Fiji: an open-source platform for biological-image analysis. Nat Methods 9, 676–682, doi: 10.1038/nmeth.2019 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ridler T & Calvard S Picture thresholding using an iterative selection method. IEEE trans syst Man Cybern 8, 630–632 (1978). [Google Scholar]
- 25.Lippincott-Schwartz J, Snapp EL & Phair RD The Development and Enhancement of FRAP as a Key Tool for Investigating Protein Dynamics. Biophys J 115, 1146–1155, doi: 10.1016/j.bpj.2018.08.007 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Borgese N, Gazzoni I, Barberi M, Colombo S & Pedrazzini E Targeting of a tail-anchored protein to endoplasmic reticulum and mitochondrial outer membrane by independent but competing pathways. Molecular Biology of the Cell 12, 2482–2496 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Omari S et al. Noncanonical autophagy at ER exit sites regulates procollagen turnover. Proceedings of the National Academy of Sciences of the United States of America 115, E10099–e10108, doi: 10.1073/pnas.1814552115 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Subach OM, Cranfill PJ, Davidson MW & Verkhusha VV An enhanced monomeric blue fluorescent protein with the high chemical stability of the chromophore. PloS one 6, e28674, doi: 10.1371/journal.pone.0028674 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Borgese N, Gazzoni I, Barberi M, Colombo S & Pedrazzini E Targeting of a tail-anchored protein to endoplasmic reticulum and mitochondrial outer membrane by independent but competing pathways. Mol Biol Cell 12, 2482–2496, doi: 10.1091/mbc.12.8.2482 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Geiser M, Cebe R, Drewello D & Schmitz R Integration of PCR fragments at any specific site within cloning vectors without the use of restriction enzymes and DNA ligase. BioTechniques 31, 88–90, 92 (2001). [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The original source data for quantification and the raw imaging data used for all the presented figures and videos will be made available from zenodo.org doi: 10.5281/zenodo.2851619.