Abstract
Tools for acute manipulation of protein localization enable elucidation of spatiotemporally defined functions, but their reliance on exogenous triggers can interfere with cell physiology. This limitation is particularly apparent for studying mitosis, whose highly choreographed events are sensitive to perturbations. Here we exploit the serendipitous discovery of a phosphorylation-controlled, cell cycle-dependent localization change of the adaptor protein PLEKHA5 to develop a system for mitosis-specific protein recruitment to the plasma membrane that requires no exogenous stimulus. Mitosis-enabled anchor-away/recruiter system comprises an engineered, 15 kDa module derived from PLEKHA5 capable of recruiting functional protein cargoes to the plasma membrane during mitosis, either through direct fusion or via GFP–GFP nanobody interaction. Applications of the mitosis-enabled anchor-away/recruiter system include both knock sideways to rapidly extract proteins from their native localizations during mitosis and conditional recruitment of lipid-metabolizing enzymes for mitosis-selective editing of plasma membrane lipid content, without the need for exogenous triggers or perturbative synchronization methods.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
Data availability
For identification of phosphopeptides, raw mass spectrometry data was processed through the Trans Proteomic Pipeline package (ver. 6.0.0) with spectral data files searched through the Comet search engine over the human UniProt proteome database downloaded on 6 November 2019. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD052237. MARS-related plasmids generated in this study have been deposited in Addgene with the following Addgene IDs: 205232, 205233, 205234, 205235, 205236, 205237, 205238 and 205239. Other data supporting the findings of this study are available within the article and the Supplementary information. Source data are provided with this paper.
References
Grecco, H. E., Schmick, M. & Bastiaens, P. I. H. Signaling from the living plasma membrane. Cell 144, 897–909 (2011).
Sunshine, H. & Iruela-Arispe, M. L. Membrane lipids and cell signaling. Curr. Opin. Lipidol. 28, 408–413 (2017).
Fegan, A., White, B., Carlson, J. C. T. & Wagner, C. R. Chemically controlled protein assembly: techniques and applications. Chem. Rev. 110, 3315–3336 (2010).
Suzuki, S. et al. A chemogenetic platform for controlling plasma membrane signaling and synthetic signal oscillation. Cell Chem. Biol. 29, 1446–1464.e10 (2022).
Kennedy, M. J. et al. Rapid blue-light–mediated induction of protein interactions in living cells. Nat. Methods 7, 973–975 (2010).
Hannanta-Anan, P., Glantz, S. T. & Chow, B. Y. Optically inducible membrane recruitment and signaling systems. Curr. Opin. Struct. Biol. 57, 84–92 (2019).
Lin, Y. C. et al. Rapidly reversible manipulation of molecular activity with dual chemical dimerizers. Angew. Chem. Int. Ed. 52, 6450–6454 (2013).
Voß, S., Klewer, L. & Wu, Y. W. Chemically induced dimerization: reversible and spatiotemporal control of protein function in cells. Curr. Opin. Chem. Biol. 28, 194–201 (2015).
Hu, J., Adebali, O., Adar, S. & Sancar, A. Dynamic maps of UV damage formation and repair for the human genome. Proc. Natl Acad. Sci. USA 114, 6758–6763 (2017).
Benman, W. et al. Temperature-responsive optogenetic probes of cell signaling. Nat. Chem. Biol. 18, 152–160 (2021).
Morgan, D. O. The Cell Cycle: Principles of Control (New Science Press Ltd, 2006).
McLean, J. R., Chaix, D., Ohi, M. D. & Gould, K. L. State of the APC/C: organization, function, and structure. Crit. Rev. Biochem. Mol. Biol. 46, 118–136 (2011).
Pomerening, J. R., Sun, Y. K. & Ferrell, J. E. Systems-level dissection of the cell-cycle oscillator: bypassing positive feedback produces damped oscillations. Cell 122, 565–578 (2005).
Sakaue-Sawano, A. et al. Genetically encoded tools for optical dissection of the mammalian cell cycle. Mol. Cell 68, 626–640.e5 (2017).
Bajar, B. T. et al. Fluorescent indicators for simultaneous reporting of all four cell cycle phases. Nat. Methods 13, 993–996 (2016).
Sakaue-Sawano, A. et al. Visualizing spatiotemporal dynamics of multicellular cell-cycle progression. Cell 132, 487–498 (2008).
Cao, X., Shami Shah, A., Sanford, E. J., Smolka, M. B. & Baskin, J. M. Proximity labeling reveals spatial regulation of the anaphase-promoting complex/cyclosome by a microtubule adaptor. ACS Chem. Biol. 17, 2605–2618 (2022).
Sluysmans, S. et al. PLEKHA5, PLEKHA6, and PLEKHA7 bind to PDZD11 to target the Menkes ATPase ATP7A to the cell periphery and regulate copper homeostasis. Mol. Biol. Cell 32, ar34 (2021).
Sluysmans, S., Méan, I., Jond, L. & Citi, S. WW, PH and C-terminal domains cooperate to direct the subcellular localizations of PLEKHA5, PLEKHA6 and PLEKHA7. Front. Cell Dev. Biol. 9, 2522 (2021).
Shami Shah, A. et al. PLEKHA4/kramer attenuates dishevelled ubiquitination to modulate wnt and planar cell polarity signaling. Cell Rep. 27, 2157–2170.e8 (2019).
Branon, T. C. et al. Efficient proximity labeling in living cells and organisms with TurboID. Nat. Biotechnol. 36, 880–887 (2018).
Kubala, M. H., Kovtun, O., Alexandrov, K. & Collins, B. M. Structural and thermodynamic analysis of the GFP:GFP–nanobody complex. Protein Sci. 19, 2389–2401 (2010).
Atilla-Gokcumen, G. E. et al. Dividing cells regulate their lipid composition and localization. Cell 156, 428 (2014).
Li, J. et al. Grp1 plays a key role in linking insulin signaling to Glut4 recycling. Dev. Cell 22, 1286–1298 (2012).
Cho, E. A. et al. Phosphorylation of RIAM by src promotes integrin activation by unmasking the PH domain of RIAM. Structure 29, 320–329.e4 (2021).
Hornbeck, P. V. et al. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 43, D512–D520 (2015).
Kinoshita, E. & Kinoshita-Kikuta, E. Improved Phos-tag SDS–PAGE under neutral pH conditions for advanced protein phosphorylation profiling. Proteomics 11, 319–323 (2011).
Johnson, J. L. et al. An atlas of substrate specificities for the human serine/threonine kinome. Nature 613, 759–766 (2023).
Bumpus, T. W. & Baskin, J. M. Clickable substrate mimics enable imaging of phospholipase d activity. ACS Cent. Sci. 3, 1070–1077 (2017).
Hardman, C. et al. Synthesis and evaluation of designed PKC modulators for enhanced cancer immunotherapy. Nat. Commun. 11, 1–11 (2020).
Gschwendt, M. et al. Inhibition of protein kinase C μ by various inhibitors. Inhibition from protein kinase c isoenzymes. FEBS Lett. 392, 77–80 (1996).
Young, L. H., Balin, B. J. & Weis, M. T. Gö 6983: a fast acting protein kinase C inhibitor that attenuates myocardial ischemia/reperfusion injury. Cardiovasc. Drug Rev. 23, 255–272 (2005).
Evenou, J. P. et al. The potent protein kinase c-selective inhibitor AEB071 (Sotrastaurin) represents a new class of immunosuppressive agents affecting early T-cell activation. J. Pharmacol. Exp. Ther. 330, 792–801 (2009).
Bibian, M. et al. Development of highly selective casein kinase 1δ/1ε (CK1δ/ε) inhibitors with potent antiproliferative properties. Bioorg. Med. Chem. Lett. 23, 4374 (2013).
Bernatík, O. et al. Functional analysis of dishevelled-3 phosphorylation identifies distinct mechanisms driven by casein kinase 1ε and Frizzled5. J. Biol. Chem. 289, 23520–23533 (2014).
Meng, Q. J. et al. Entrainment of disrupted circadian behavior through inhibition of casein kinase 1 (CK1) enzymes. Proc. Natl Acad. Sci. USA 107, 15240–15245 (2010).
Rena, G., Bain, J., Elliott, M. & Cohen, P. D4476, a cell-permeant inhibitor of CK1, suppresses the site-specific phosphorylation and nuclear exclusion of FOXO1a. EMBO Rep. 5, 60–65 (2004).
Moura, M. & Conde, C. Phosphatases in mitosis: roles and regulation. Biomolecules 9, 55 (2019).
Kastian, R. F. et al. Dephosphorylation of neural wiring protein shootin1 by PP1 phosphatase regulates netrin-1-induced axon guidance. J. Biol. Chem. 299, 104687 (2023).
Swingle, M., Ni, L. & Honkanen, R. E. Small-molecule inhibitors of Ser/Thr protein phosphatases. Methods Mol. Biol. 365, 23–38 (2007).
Bastan, R., Eskandari, N., Ardakani, H. J. & Peachell, P. T. Effects of fostriecin on β2-adrenoceptor-driven responses in human mast cells. J. Immunotoxicol. 14, 60–65 (2017).
McKinley, K. L. & Cheeseman, I. M. Polo-like kinase 1 licenses CENP-a deposition at centromeres. Cell 158, 397–411 (2014).
Selvy, P. E., Lavieri, R. R., Lindsley, C. W. & Brown, H. A. Phospholipase D: enzymology, functionality, and chemical modulation. Chem. Rev. 111, 6064–6119 (2011).
Tei, R. & Baskin, J. M. Spatiotemporal control of phosphatidic acid signaling with optogenetic, engineered phospholipase Ds. J. Cell Biol. 219, e201907013 (2020).
Tei, R., Bagde, S. R., Fromme, J. C. & Baskin, J. M. Activity-based directed evolution of a membrane editor in mammalian cells. Nat. Chem. 15, 1030–1039 (2023).
Balla, T. Phosphoinositides: tiny lipids with giant impact on cell regulation. Physiol. Rev. 93, 1019–1137 (2013).
Idevall-Hagren, O., Dickson, E. J., Hille, B., Toomre, D. K. & De Camilli, P. Optogenetic control of phosphoinositide metabolism. Proc. Natl Acad. Sci. USA 109, E2316–E2323 (2012).
Cauvin, C. & Echard, A. Phosphoinositides: lipids with informative heads and mastermind functions in cell division. Biochim. Biophys. Acta 1851, 832–843 (2015).
Toyoshima, F., Matsumura, S., Morimoto, H., Mitsushima, M. & Nishida, E. PtdIns(3,4,5)P3 regulates spindle orientation in adherent cells. Dev. Cell 13, 796–811 (2007).
Kotak, S., Busso, C. & Gönczy, P. NuMA interacts with phosphoinositides and links the mitotic spindle with the plasma membrane. EMBO J. 33, 1815–1830 (2014).
Bordhan, P., Razavi Bazaz, S., Jin, D. & Ebrahimi Warkiani, M. Advances and enabling technologies for phase-specific cell cycle synchronisation. Lab Chip 22, 445–462 (2022).
Fiume, R. et al. Involvement of nuclear PLCβl in lamin B1 phosphorylation and G 2 /M cell cycle progression. FASEB J. 23, 957–966 (2009).
Goss, V. L. et al. Identification of nuclear pII protein kinase C as a mitotic lamin kinase. J. Biol. Chem. 269, 19074–19080 (1994).
Sun, B., Murray, N. R. & Fields, A. P. A role for nuclear phosphatidylinositol-specific phospholipase C in the G2/M phase transition. J. Biol. Chem. 272, 26313–26317 (1997).
Lim, S. et al. BioPROTACs as versatile modulators of intracellular therapeutic targets including proliferating cell nuclear antigen (PCNA). Proc. Natl Acad. Sci. USA 117, 5791–5800 (2020).
Ludwicki, M. B. et al. Broad-spectrum proteome editing with an engineered bacterial ubiquitin ligase mimic. ACS Cent. Sci. 5, 852–866 (2019).
Siriwardena, S. U. et al. Phosphorylation-inducing chimeric small molecules. J. Am. Chem. Soc. 142, 14052–14057 (2020).
Chen, P. H. et al. Modulation of phosphoprotein activity by phosphorylation targeting chimeras (PhosTACs). ACS Chem. Biol. 16, 2808–2815 (2021).
Ramirez, D. H. et al. Engineering a proximity-directed O-GlcNAc transferase for selective protein O-GlcNAcylation in cells. ACS Chem. Biol. 15, 1059–1066 (2020).
Ge, Y. et al. Target protein deglycosylation in living cells by a nanobody-fused split O-GlcNAcase. Nat. Chem. Biol. 17, 593–600 (2021).
Wang, W. et al. A light- and calcium-gated transcription factor for imaging and manipulating activated neurons. Nat. Biotechnol. 35, 864–871 (2017).
Barnea, G. et al. The genetic design of signaling cascades to record receptor activation. Proc. Natl Acad. Sci. USA 105, 64–69 (2008).
Kim, M. W. et al. Time-gated detection of protein-protein interactions with transcriptional readout. eLife 6, e30233 (2017).
Gao, X. J., Chong, L. S., Kim, M. S. & Elowitz, M. B. Programmable protein circuits in living cells. Science 361, 1252–1258 (2018).
Fink, T. et al. Design of fast proteolysis-based signaling and logic circuits in mammalian cells. Nat. Chem. Biol. 15, 115–122 (2018).
Yuan, J. et al. Multi-responsive self-healing metallo-supramolecular gels based on ‘click’ ligand. J. Mater. Chem. 22, 11515–11522 (2012).
Alamudi, S. H. et al. Development of background-free tame fluorescent probes for intracellular live cell imaging. Nat. Commun. 7, 1–9 (2016).
Shami Shah, A., Cao, X., White, A. C. & Baskin, J. M. PLEKHA4 promotes Wnt/b-catenin signaling-mediated G 1-S transition and proliferation in melanoma. Cancer Res. 81, 2029–2043 (2021).
Cao, X. & Baskin, J. M. Applying the mitosis-enabled anchor-away/recruiter system (MARS) for conditional protein recruitment to the plasma membrane during mitosis. https://doi.org/10.17504/protocols.io.n2bvjnzypgk5/v1 (2024).
McIntire, L. B. J. et al. Reduction of synaptojanin 1 ameliorates synaptic and behavioral impairments in a mouse model of alzheimer’s disease. J. Neurosci. 32, 15271–15276 (2012).
Nasuhoglu, C. et al. Nonradioactive analysis of phosphatidylinositides and other anionic phospholipids by anion-exchange high-performance liquid chromatography with suppressed conductivity detection. Anal. Biochem. 301, 243–254 (2002).
Deutsch, E. W. et al. Trans-proteomic pipeline: robust mass spectrometry-based proteomics data analysis suite. J. Proteome Res. 22, 615–624 (2023).
Eng, J. K., Jahan, T. A. & Hoopmann, M. R. Comet: an open-source MS/MS sequence database search tool. Proteomics 13, 22–24 (2013).
Perez-Riverol, Y. et al. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 50, D543–D552 (2022).
Nagy, Z., Comer, S. & Smolenski, A. Analysis of protein phosphorylation using phos-tag gels. Curr. Protoc. Protein Sci. 93, e64 (2018).
Acknowledgements
We acknowledge support from the National Institutes of Health (NIH) grant R01GM143367 (J.M.B.), NIH grant T32GM138826 (S.H.), NIH grant R35GM141159 (M.B.S.), the Sloan Research Fellowship (J.M.B.) and the DoD DURIP GRANT13369767 and GRANT13710486 (L.B.M.). We thank I. Cheeseman for valuable discussions and for providing the PLK1–GFP cell line, and we thank L. Cantley for sharing the kinase predictions of the PLEKHA5 S161 site. We thank R. Tei, J. Li and N. Vasireddi for technical assistance, the Bretscher lab for the HEK 293TN cell line, the Lammerding lab for the pCDH-CMV-MCS-EF1α-Puro, PAX2 and VSVg plasmids, and access to the IncuCyte system, the Fromme and Emr labs for equipment, and members of the Baskin lab for helpful discussions.
Author information
Authors and Affiliations
Contributions
Conceptualization: X.C. and J.M.B. Funding acquisition: L.B.M., M.B.S. and J.M.B. Investigation: X.C., S.H., M.M.W., Y.-T.C., D.-C.C., K.M.W. and A.L. Project administration: L.B.M., M.B.S. and J.M.B. Supervision: L.B.M., M.B.S. and J.M.B. Writing—original draft: X.C. and J.M.B. Writing—review and editing: X.C., S.H., M.M.W., Y.-T.C., D.-C.C., L.B.M., M.B.S. and J.M.B.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Cell Biology thanks Raymond Blind, Haibo Jiang, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 A pool of PLEKHA5 translocates to the PM during mitosis by binding to PI(4,5)P2.
(a) Live-cell confocal microscopy of HeLa cells transiently expressing GFP-tagged PLEKHA5 constructs in interphase and mitosis. Imaging experiment was repeated three times independently with similar results. (b) Representative series of still images from a time-lapse movie in asynchronous, stable H2B-mCherry-expressing (magenta) HeLa cells transfected with PLEKHA5WW-H-BP-PH-GFP (green). Numbers indicate time before/after nuclear envelope breakdown (defined as t = 0 min). Imaging experiment was repeated three times independently with similar results. (c) Schematic representation of proximity biotinylation strategy using Lyn10-TurboID-V5 to biotinylate PM-proximal proteins. (d) Lyn10-TurboID showed expected PM-specific localization and biotinylation activity as demonstrated by staining for V5 (green) and biotin (streptavidin-Alexa Fluor 568, magenta). Imaging experiment was repeated three times independently with similar results. (e) Western blot analysis confirmed that Lyn10-TurboID biotinylates a protein marker of the PM but not markers of other subcellular localizations. The experiment was repeated two times independently with similar results. (f) Clustal Omega alignment of the primary amino acid sequences of the PH domain of PLEKHA4 and PLEKHA5. Highlighted in red are two arginine residues previously established to be required for binding PI(4,5)P2 in PLEKHA4. (g) Confocal microscopy of HeLa cells transfected with the R190A and R244 A mutants of PLEKHA5WW-H-BP-PH-GFP and PLEKHA5H-BP-PH-GFP. Imaging experiment was repeated two times independently with similar results. Unprocessed blots are available in source data.
Extended Data Fig. 2 Design and engineering of MARS and MARS-nGFP.
(a) Clustal Omega alignment of primary amino acid sequences around the ‘4A’ region of PLEKHA4 and PLEKHA5. (b) Confocal microscopy of the K157A and R158A mutants of PLEKHA5H-BP-PH-GFP. Imaging experiment was repeated two times independently with similar results. (c, d) Representative series of still images from time-lapse movies in asynchronous HeLa cells stably expressing H2B-tagBFP and transfected with either MARS (c) or 2xMARS-nGFP (d). MARS constructs are colored magenta and H2B-tagBFP is colored green. Numbers indicate time before/after nuclear envelope breakdown (defined as t = 0 min). Each imaging experiment was repeated two times independently with similar results.
Extended Data Fig. 3 Characterization of stoichiometry of 2xMARS-nGFP and effects of MARS systems on cell proliferation rates.
(a) Four stable cell lines were generated via lentiviral transduction that express similar levels of 2xMARS-nGFP and varying levels of GFP, which was under control of either a PGK or CMV promoter (the three CMV-GFP-expressing lines were isolated by FACS, sorting for different levels of GFP expression). Shown are representative Western blots showing GFP (asterisk denotes a longer CMV-GFP product potentially arising from an upstream alternative start codon) and 2xMARS-nGFP (mCherry blot) expression levels in these cell lines alongside blots of purified GFP and mCherry protein standards. (b) Quantification of GFP to 2xMARS-nGFP molar ratios in HeLa cells stably expressing 2xMARS-nGFP and various levels of GFP from (A) (n = 3 biological replicates, one-way ANOVA with Tukey post hoc multiple comparisons test). (c, d) Imaging analysis of GFP recruitment to the PM during mitosis at different stoichiometries of GFP to 2xMARS-nGFP determined by quantitative image analysis. (c) Quantification of the PM to cytosolic fluorescence ratios of GFP as a readout for measuring GFP recruitment levels by 2xMARS-nGFP. For PGK-GFP: n = 8 cells for low GFP expression, n = 4 cells for medium expression, and n = 7 cells for high expression. For CMV-GFP cell lines, n = 10 cells each for low, medium, and high GFP expression level. Cells analyzed came from two independent experiments (one-way ANOVA with Tukey post hoc multiple comparisons test). Note: CMV-GFP cells were sorted by FACS to generate three separate cell lines with low, medium, and high GFP expression, and images were acquired for each cell line. For PGK-GFP cells, the low, medium, and high cells were all from a single cell line and are categorized based on GFP fluorescence intensity during imaging analysis by ImageJ post-acquisition. (d) Representative micrographs for HeLa cells stably expressing 2xMARS-nGFP and varying levels of GFP analyzed in (c). Brightness of the GFP channel was adjusted post-acquisition for better visualization of the weaker fluorescence signals for PGK-GFP and CMV-GFP low cells. (e) Quantification of mean fluorescence intensity in the cytosol during interphase as a means to determine GFP expression levels in PGK-GFP low, medium, high cells and CMV-GFP low cells (n = 40 cells from two independent experiments). Compared to CMV-GFP low cells, PGK-GFP low cells exhibited an average of 7.6x lower expression, PGK-GFP medium cells exhibited ~2.2x lower expression, and PGK-GFP-high cells exhibited ~1.1x higher expression. Converting to GFP:2xMARS-nGFP molar ratio in these cells, the estimated ratios are 1.8 (low), 6.2 (medium), and 15.1 (high). (f, g) Ectopic expression of MARS and 2xMARS-nGFP did not negatively impact rates of cell division and proliferation. (f) Representative growth curves for wild-type (WT) HeLa cells and those stably expressing MARS and 2xMARS-nGFP (n = 2 technical replicates for each curve). Raw data (faded-color curves with error bar) acquired by IncuCyte Live-Cell Analysis System were fitted using GraphPad Prism (Exponential growth curve model). Fitted curves are shown as solid lines. (g) Doubling time of the three cell lines calculated from the fitted growth curves (n = 3 biological replicates, one-way ANOVA with Tukey post hoc multiple comparisons test). Source numerical data and unprocessed blots are available in Source data.
Extended Data Fig. 4 Mutational analysis reveals no contributions of phosphorylation of residues other than S161 to the cell cycle-dependent localization of PLEKHA5.
Live-cell confocal microscopy of phosphodeficient (Ser/Thr to Ala and Tyr to Phe) and phosphomimetic (Ser/Thr to Asp and Tyr to Glu) mutants of GFP-tagged PLEKHA5 full-length protein (a) or WW-H-BP-PH fragment (b). Imaging experiment was repeated two times independently with similar results.
Extended Data Fig. 5 S161 phosphorylation status influences the PM localization of EGFP-PLEKHA5FL and PLEKHA5WW-H-BP-PH-EGFP.
(a, b) Live-cell confocal microscopy of the subcellular localizations of the WT, S161A, and S161D variants of full-length PLEKHA5FL (a) and the WW-H-BP-PH motifs (b). (c, d) Quantification of PM localization for GFP-PLEKHA5FL (c) and PLEKHA5WW-H-BP-PH-GFP (d) using colocalization analysis. Pearson coefficients were computed from images acquired in HeLa cells transiently expressing either of the PLEKHA5 constructs with a transfectable PM marker, Lyn10-LOV-mCherry. For GFP-PLEKHA5FL in interphase: n = 37 cells for WT, n = 35 cells for S161A, and n = 30 cells for S161D; in mitosis: n = 12 cells for WT and S161D and n = 11 cells for S161A. For PLEKHA5WW-H-BP-PH-GFP in interphase, n = 26 cells for WT, n = 29 cells for S161A, and n = 25 cells for S161D; in mitosis: n = 13 cells for WT and S161D and n = 16 cells for S161A. Cells analyzed came from three independent experiments (one-way ANOVA with Tukey post hoc multiple comparisons test). (e, f) Quantification of PM association for transfected GFP-PLEKHA5FL (e) and PLEKHA5WW-H-BP-PH-GFP (f) using PM-targeted proximity biotinylation. Representative western blots are shown at top with quantification of GFP signals in the streptavidin pulldowns at bottom (n = 4 biological replicates, one-way ANOVA with Tukey post hoc multiple comparisons test). Source numerical data and unprocessed blots are available in Source data.
Extended Data Fig. 6 Requirement for PI(4,5)P2 binding in S161-mediated PM localization in additional PLEKHA5 constructs and conservation of S161-mediated PM localization in PLEKHA4.
(a, b) Live-cell confocal microscopy of HeLa cells transiently expressing combinational mutants of S161A with either R190A or R244A in PLEKHA5FL-GFP (a) and PLEKHA5H-BP-PH-GFP (b). Imaging experiment was repeated two times independently with similar results. (c) Confocal microscopy of HeLa cells transfected with either the WT, A46S, or A46D forms of full-length PLEKHA4. Imaging experiment was repeated two times independently with similar results. (d) Clustal Omega alignment of amino acid sequences around A46 in PLEKHA4 and S161 in PLEKHA5.
Extended Data Fig. 7 PKC and CK1 kinase activities contribute to S161 phosphorylation.
(a, b) PLEKHA5WW-H-BP-PH-GFP displayed reduced PM localization upon activation of PKC by 100 nM PMA (a) or 200 nM Bryostatin 1 (b). Representative micrographs before and after agonist treatment for each condition are shown at left, with quantifications of the ratios of PM to cytosolic fluorescence (excluding microtubule signal) before and after treatment plotted on the right (n = 12 cells from three independent experiments, one-way ANOVA with Sidak post hoc multiple comparisons test). (c) Top 10 kinase candidates for phosphorylating S161 (highlighted in red) if S159 serves as the priming phosphorylation site. (d) Inhibition of CK1 activity by 5 µM PF670462 or 100 µM D4476 decreased S161 phosphorylation. Shown are representative western blots and quantification for Phos-tag and regular SDS-PAGE analysis of PLEKHA5H-BP-PH-GFP (n = 3 biological replicates, one-way ANOVA with Tukey post hoc multiple comparisons test). (e) Phos-tag SDS-PAGE analysis of S161 phosphorylation upon treatment of inhibitors of the PPP family of protein Ser/Thr phosphatases (Tautomycetin, Fostriecin, and Okadaic acid, each at 1 µM), probing with GFP and phospho-Thr antibodies. The experiment was repeated two times independently with similar results. (f) RT-qPCR analysis of knockdown efficiency for siRNAs targeting CK1 isoforms (CSNK1A1, CSNK1D, CSNK1E), PKC isoforms (PRKCA, PRKCD), and PPP catalytic subunits (PPP1CA, PPP1CB, PPP2CA). (g) Knockdown of CK1 (triple knockdown of CK1a, CK1d, and CK1e) and PKC (double knockdown of PKCa and PKCd) decreased S161 phosphorylation, whereas depletion of PP1 and PP2A (triple knockdown of two PP1catalytic subunits and one PP2A catalytic subunit) increased S161 phosphorylation. Shown are representative Western blots and quantification for Phos-tag and regular SDS-PAGE analysis of PLEKHA5H-BP-PH-GFP (n = 3 biological replicates, unpaired, two-tailed Student’s t-test). Source numerical data and unprocessed blots are available in Source data.
Extended Data Fig. 8 Evaluation of PM localization of additional MARS constructs for PLD and PI3K and time-lapse imaging of MARS and MARS-p85iSH2 during mitosis.
(a) Live-cell confocal microscopy of HeLa cells transfected with the single MARS fusion to WT or catalytic dead (H170A) forms of PLDPMF revealing only partial recruitment to the PM during mitosis. (b) Representative confocal micrographs of HeLa cells transfected with 2xMARS-nGFP (magenta) and H170A mutant of GFP-PLDPMF (green) showing effective PM recruitment only during mitosis. (c) Live-cell confocal microscopy of HeLa cells transiently expressing the 2xMARS-p85iSH2 construct, revealing undesired partial PM localization during interphase. (d, e) Representative still images from time-lapse movies in asynchronous HeLa cells stably expressing H2B-tagBFP and transfected with either MARS (d) or MARS-p85iSH2 (e), and the PI(3,4,5)P3 marker GFP-AktPH. Images display merged channels of H2B-tagBFP (cyan) and MARS constructs (magenta). Numbers indicate time before/after nuclear envelope breakdown (defined as t = 0 min). All imaging experiments were repeated two times independently with similar results.
Supplementary information
Supplementary Information
Legends for Supplementary Table 1 and Supplementary Videos 1–8. Supplementary Figs. 1–4 and representative gating strategies for flow cytometry experiments.
Supplementary Table 1
siRNA duplex sequences and RT–qPCR primer sequences.
Supplementary Video 1
(Related to Extended Data Fig. 1b) Time-lapse confocal microscopy of asynchronous HeLa cells stably expressing H2B-mCherry and transiently transfected with PLEKHA5WW-H-BP-PH–GFP, during mitosis, with cells incubated at 37 °C and images acquired every 1 min. Left: H2B-mCherry. Middle: PLEKHA5WW-H-BP-PH–GFP. Right: merge (mCherry, magenta; GFP, green).
Supplementary Video 2
(Related to Extended Data Fig. 2c) Time-lapse confocal microscopy of asynchronous HeLa cells stably expressing H2B-tagBFP and transiently transfected with MARS, during mitosis, with cells incubated at 37 °C and images acquired every 2 min. Left: H2B-tagBFP. Middle: MARS. Right: merge (tagBFP, green; MARS, magenta).
Supplementary Video 3
(Related to Extended Data Fig. 2d) Time-lapse confocal microscopy of asynchronous HeLa cells stably expressing H2B-tagBFP and transiently transfected with 2xMARS–nGFP, during mitosis, with cells incubated at 37 °C and images acquired every 2 min. Left: H2B-tagBFP. Middle: MARS. Right: merge (tagBFP, green; 2xMARS–nGFP, magenta).
Supplementary Video 4
(Related to Extended Data Fig. 3e) Time-lapse video acquired by the IncuCyte Live-Cell Analysis System of WT HeLa cells incubated at 37 °C with 5% CO2. Images were acquired every 15 min.
Supplementary Video 5
(Related to Extended Data Fig. 3e) Time-lapse video acquired by the IncuCyte Live-Cell Analysis System of HeLa cells stably expressing MARS, with cells incubated at 37 °C with 5% CO2 and images acquired every 15 min.
Supplementary Video 6
(Related to Extended Data Fig. 3e) Time-lapse video acquired by the IncuCyte Live-Cell Analysis System of HeLa cells stably expressing 2xMARS–nGFP, with cells incubated at 37 °C with 5% CO2 and images acquired every 15 min.
Supplementary Video 7
(Related to Fig. 6e and Extended Data Fig. 8d) Time-lapse confocal microscopy of asynchronous HeLa cells stably expressing H2B-tagBFP and transiently transfected with MARS and the PI(3,4,5)P3 marker GFP-AktPH, during mitosis, with cells incubated at 37 °C and images acquired every 4 min. The left video displays merged channels of H2B-tagBFP (cyan) and MARS (magenta), and the right video displays merged channels of H2B-tagBFP (cyan) and GFP-AktPH (yellow).
Supplementary Video 8
(Related to Fig. 6f and Extended Data Fig. 8e) Time-lapse confocal microscopy of asynchronous HeLa cells stably expressing H2B-tagBFP and transiently transfected with MARS–p85iSH2 and the PI(3,4,5)P3 marker GFP-AktPH, during mitosis, with cells incubated at 37 °C and images acquired every 4 min. The left video displays merged channels of H2B-tagBFP (cyan) and MARS–p85iSH2 (magenta), and the right video displays merged channels of H2B-tagBFP (cyan) and GFP-AktPH (yellow).
Source data
Source Data Fig. 1
Statistical source data.
Source Data Figs. 1, 3 and 4 and Extended Data Figs. 1, 3, 5 and 7.
Unprocessed western blots.
Source Data Fig. 3
Statistical source data.
Source Data Fig. 4
Statistical source data.
Source Data Fig. 5
Statistical source data.
Source Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 3/Table 3
Statistical source data.
Source Data Extended Data Fig. 5/Table 5
Statistical source data.
Source Data Extended Data Fig. 7/Table 7
Statistical source data.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Cao, X., Huang, S., Wagner, M.M. et al. A phosphorylation-controlled switch confers cell cycle-dependent protein relocalization. Nat Cell Biol (2024). https://doi.org/10.1038/s41556-024-01495-8
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41556-024-01495-8