Volume 27, Issue 24, Pages e8 (December 2017)

Slides:



Advertisements
Similar presentations
Harinath Doodhi, Eugene A. Katrukha, Lukas C. Kapitein, Anna Akhmanova 
Advertisements

Volume 24, Issue 19, Pages (October 2014)
Harinath Doodhi, Eugene A. Katrukha, Lukas C. Kapitein, Anna Akhmanova 
Volume 21, Issue 2, Pages (January 2011)
Volume 126, Issue 4, Pages (August 2006)
DELLAs Modulate Jasmonate Signaling via Competitive Binding to JAZs
Volume 19, Issue 23, Pages (December 2009)
Volume 17, Issue 24, Pages (December 2007)
Structure of the Papillomavirus DNA-Tethering Complex E2:Brd4 and a Peptide that Ablates HPV Chromosomal Association  Eric A. Abbate, Christian Voitenleitner,
Volume 40, Issue 4, Pages (November 2010)
Volume 16, Issue 6, Pages (December 2004)
Volume 20, Issue 7, Pages (April 2010)
NRF2 Is a Major Target of ARF in p53-Independent Tumor Suppression
Volume 24, Issue 19, Pages (October 2014)
Volume 24, Issue 15, Pages (August 2014)
Volume 26, Issue 2, Pages (January 2016)
Volume 15, Issue 22, Pages (November 2005)
Sherilyn Grill, Valerie M. Tesmer, Jayakrishnan Nandakumar 
Communication with the Exon-Junction Complex and Activation of Nonsense-Mediated Decay by Human Upf Proteins Occur in the Cytoplasm  Guramrit Singh, Steffen.
Volume 23, Issue 3, Pages (February 2013)
Elif Nur Firat-Karalar, Navin Rauniyar, John R. Yates, Tim Stearns 
Volume 14, Issue 1, Pages (January 2004)
The Intracellular Domain of the Frazzled/DCC Receptor Is a Transcription Factor Required for Commissural Axon Guidance  Alexandra Neuhaus-Follini, Greg J.
Volume 14, Issue 10, Pages (October 2007)
Volume 30, Issue 3, Pages (August 2014)
Beena Krishnan, Lila M. Gierasch  Chemistry & Biology 
EB3 Regulates Microtubule Dynamics at the Cell Cortex and Is Required for Myoblast Elongation and Fusion  Anne Straube, Andreas Merdes  Current Biology 
Wood Cell-Wall Structure Requires Local 2D-Microtubule Disassembly by a Novel Plasma Membrane-Anchored Protein  Yoshihisa Oda, Yuki Iida, Yuki Kondo,
Vanessa Brès, Tomonori Yoshida, Loni Pickle, Katherine A. Jones 
Volume 19, Issue 13, Pages (July 2009)
CENP-C Is a Structural Platform for Kinetochore Assembly
Maïlys A.S. Vergnolle, Stephen S. Taylor  Current Biology 
Architecture Dependence of Actin Filament Network Disassembly
Volume 22, Issue 20, Pages (October 2012)
Functional Comparison of H1 Histones in Xenopus Reveals Isoform-Specific Regulation by Cdk1 and RanGTP  Benjamin S. Freedman, Rebecca Heald  Current Biology 
The Role of NEDD1 Phosphorylation by Aurora A in Chromosomal Microtubule Nucleation and Spindle Function  Roser Pinyol, Jacopo Scrofani, Isabelle Vernos 
Volume 23, Issue 8, Pages (May 2018)
Volume 11, Issue 21, Pages (October 2001)
Lizhong Xu, Veronica Lubkov, Laura J. Taylor, Dafna Bar-Sagi 
Volume 21, Issue 15, Pages (August 2011)
Volume 42, Issue 2, Pages e3 (July 2017)
Geoffrey J. Guimaraes, Yimin Dong, Bruce F. McEwen, Jennifer G. DeLuca 
Codependent Activators Direct Myoblast-Specific MyoD Transcription
Gloria Slattum, Yapeng Gu, Roger Sabbadini, Jody Rosenblatt 
Volume 26, Issue 1, Pages e4 (January 2018)
Myosin-IXA Regulates Collective Epithelial Cell Migration by Targeting RhoGAP Activity to Cell-Cell Junctions  Tatiana Omelchenko, Alan Hall  Current.
Minus-End-Directed Motor Ncd Exhibits Processive Movement that Is Enhanced by Microtubule Bundling In Vitro  Ken'ya Furuta, Yoko Yano Toyoshima  Current.
Volume 16, Issue 14, Pages (July 2006)
Volume 21, Issue 2, Pages (January 2011)
Volume 17, Issue 8, Pages (April 2007)
Physcomitrella patens Auxin-Resistant Mutants Affect Conserved Elements of an Auxin- Signaling Pathway  Michael J. Prigge, Meirav Lavy, Neil W. Ashton,
Volume 21, Issue 15, Pages (August 2011)
ASPP2 Regulates Epithelial Cell Polarity through the PAR Complex
Kari Barlan, Wen Lu, Vladimir I. Gelfand  Current Biology 
Volume 129, Issue 2, Pages (April 2007)
Volume 17, Issue 18, Pages (September 2007)
Volume 21, Issue 12, Pages (June 2011)
Wood Cell-Wall Structure Requires Local 2D-Microtubule Disassembly by a Novel Plasma Membrane-Anchored Protein  Yoshihisa Oda, Yuki Iida, Yuki Kondo,
Wenxiang Meng, Yoshimi Mushika, Tetsuo Ichii, Masatoshi Takeichi  Cell 
Volume 15, Issue 14, Pages (July 2005)
Uma B. Karadge, Minja Gosto, Matthew L. Nicotra  Current Biology 
Structure of the Siz/PIAS SUMO E3 Ligase Siz1 and Determinants Required for SUMO Modification of PCNA  Ali A. Yunus, Christopher D. Lima  Molecular Cell 
Growth Factor-Dependent Trafficking of Cerebellar NMDA Receptors via Protein Kinase B/Akt Phosphorylation of NR2C  Bo-Shiun Chen, Katherine W. Roche 
In Vitro Analysis of Huntingtin-Mediated Transcriptional Repression Reveals Multiple Transcription Factor Targets  Weiguo Zhai, Hyunkyung Jeong, Libin.
Minus-End-Directed Motor Ncd Exhibits Processive Movement that Is Enhanced by Microtubule Bundling In Vitro  Ken'ya Furuta, Yoko Yano Toyoshima  Current.
Volume 9, Issue 1, Pages (January 2002)
Ricksen S. Winardhi, Qingnan Tang, Jin Chen, Mingxi Yao, Jie Yan 
Volume 65, Issue 5, Pages e4 (March 2017)
Zackary N. Scholl, Weitao Yang, Piotr E. Marszalek  Biophysical Journal 
Volume 33, Issue 3, Pages (May 2015)
Presentation transcript:

Volume 27, Issue 24, Pages 3783-3795.e8 (December 2017) Tension-Dependent Stretching Activates ZO-1 to Control the Junctional Localization of Its Interactors  Domenica Spadaro, Shimin Le, Thierry Laroche, Isabelle Mean, Lionel Jond, Jie Yan, Sandra Citi  Current Biology  Volume 27, Issue 24, Pages 3783-3795.e8 (December 2017) DOI: 10.1016/j.cub.2017.11.014 Copyright © 2017 Elsevier Ltd Terms and Conditions

Current Biology 2017 27, 3783-3795.e8DOI: (10.1016/j.cub.2017.11.014) Copyright © 2017 Elsevier Ltd Terms and Conditions

Figure 1 Stretched and Folded and/or Disordered ZO-1 Conformations Are Detected by SIM and PLA (A) Scheme of myc-ZO-1-HA expressed in ZO-1-KO Eph4 cells. (B–G) SIM images of cells expressing exogenous myc-ZO-1-HA, and treated either with control siRNA either in the absence (B, F, and G) or in the presence (C) of blebbistatin, or with si-ZO-2 either in the absence (D and F’) or in the presence (E and F”) of blebbistatin. Red (Cy3) and green (Alexa 488) fluorophores (insets/arrows) label N- and C-terminal ends of ZO-1. Yellow arrows indicate overlapped signal. Experimental treatments (siRNA and blebbistatin) are indicated on the left of images. (F) si-control, (F’) si-ZO-2, and (F”) si-ZO2+blebbistatin show calibration of chromatic shift, where junctions are imaged next to TetraSpeck Fluorescent microspheres (b, beads; merge image in bottom right panel). (G) The junction between two cells expressing tagged ZO-1. (H) Distribution of fluorophores/tags signal intensities as a function of distance. (I) Distance between intensity peaks. Error bars indicate SD. (J and K) PLA analysis on cells expressing myc-ZO-1HA, with the indicated treatments. Arrows and arrowheads indicate strong or weak PLA signal, respectively. The scale bars represent 5 μm (B–E) and 30 μm (J and K). Related Figure S1 shows experimental system, control experiments, and additional measures and quantifications. Related Figure S7 shows schemes of ZO-1 in stretched and folded conformations under the different experimental conditions. Current Biology 2017 27, 3783-3795.e8DOI: (10.1016/j.cub.2017.11.014) Copyright © 2017 Elsevier Ltd Terms and Conditions

Figure 2 Mechanical Stretching of Single Full-Length ZO-1 Molecules In Vitro (A) Scheme of experimental setup. (B) Force-extension curves of ZO-1 unfolding (colored) and refolding (gray). (C) Zoom-in of refolding events during force-decrease scans (loading rate = −0.1 pN/s) from (B). (D) Distribution of the ZO-1 domains unfolding forces (loading rate = 1 pN/s). (E) Distribution of number of residues involved in the individual unfolding events (N, number of unfolding events). The inset shows the distribution of total number of residues involved in unfolding in individual force-increase scans (N, number of scans). (F) Theoretical calculation of the force-extension curves of full-length ZO-1. Numbers of residues assumed to be non-structured are indicated. Related Figure S2 shows additional extension/refolding curves and step size measurements. Current Biology 2017 27, 3783-3795.e8DOI: (10.1016/j.cub.2017.11.014) Copyright © 2017 Elsevier Ltd Terms and Conditions

Figure 3 The Junctional Accumulation and Expression of DbpA Are Regulated by Force in a ZO-Dependent Manner (A, B, E, and F) Immunofluorescent localization of DbpA, ZO-2, PLEKHA7 (internal reference for junctions), and ZO-1 either in WT (A and B) or mixed WT+ZO-1-KO (E and F) cells following treatment with either si-control or si-ZO-2, with the indicated drugs treatment. The scale bar represents 5 μm. The localization of occludin is shown in related Figure S3. Arrows and arrowheads indicate normal or decreased/undetected junctional labeling. (n) in (F) (BL/MG) indicates nuclear labeling for DbpA. Merge images show nuclei in blue (DAPI). (C and D) Quantification of junctional labeling of ZO-2 and DbpA (C) and immunoblotting analysis of the indicated proteins following experimental treatments (D). (G and H) Immunoblot of the indicated junctional proteins (red and green boxes indicate decreased and rescued DbpA; G) and DbpA levels quantification following indicated experimental treatments (H). Asterisks indicate depleted/KO cells in immunofluorescence and statistical significance in plots. Error bars indicate SD. Current Biology 2017 27, 3783-3795.e8DOI: (10.1016/j.cub.2017.11.014) Copyright © 2017 Elsevier Ltd Terms and Conditions

Figure 4 Tension Controls Cell Proliferation and Paracellular Barrier Function through ZO Proteins (A) Analysis of proliferation (% Ki67 positivity) of cells grown in 2D, with the indicated treatments. NT, not treated. (B and B’) Immunoblotting analysis (B) and quantification of DbpA levels (B’). (C and D) mRNA levels (relative expression by qRT-PCR, 100% = synchronized proliferating cells in G1) of DbpA target genes Cyclin D1 and PCNA (C) and ErbB2 (D) following the indicated experimental treatments. (E and F) Transepithelial electrical resistance (TEER) (E) and dextran permeability (Papp) in WT and ZO-1-KO cells (F), with the indicated treatments. Asterisks indicate statistical significance. Error bars indicate SD. Related Figure S7 shows schematically the different fates of DbpA and its activity on target genes, depending on the experimental conditions. Current Biology 2017 27, 3783-3795.e8DOI: (10.1016/j.cub.2017.11.014) Copyright © 2017 Elsevier Ltd Terms and Conditions

Figure 5 ZO Proteins and Actomyosin Contractility Control Cell Proliferation, Cyst Growth, and DbpA Localization in Eph4 Cells Grown in Matrigel (A–D) Brightfield microscopy images of Eph4 cysts (A and B, WT; C and D, ZO-1-KO) at different days (d) of culture in Matrigel, without (A and C) or with (B and D) dATP treatment. Asterisks at the 21d time point indicate cyst lumens. Scale bars represent 10 μm. (E) Analysis of cyst size (diameter). Cell proliferation/number in cysts is shown in related Figure S4. Error bars indicate SD. (F) Immunoblotting analysis of levels of the indicated proteins in lysates of cysts at 21d. Red/green box indicates decreased/rescued DbpA levels. (G–J) Immunofluorescent localization of DbpA and E-cadherin (to label junctions) in untreated or dATP-treated (dATP) WT or ZO-1-KO cysts at days 4 (G and I) and 21 (H and J). Arrows and arrowheads indicate normal and reduced/undetected staining, respectively. Current Biology 2017 27, 3783-3795.e8DOI: (10.1016/j.cub.2017.11.014) Copyright © 2017 Elsevier Ltd Terms and Conditions

Figure 6 Intra-molecular Interactions between ZPSG and C-Terminal Domains of ZO Proteins Prevent Binding of DbpA (A and C) Schemes of ZO-1 (A) and ZO-2 (C), with the indicated domains, and prey (GFP-tag/green) and bait (GST-tag/red) constructs for pull-down experiments. (B, B’, and D) Interaction between ZPSG and C-terminal regions of ZO-1 (B; quantification in B’, with error bars indicating SD), and ZO-2 (D). (E–J) Competition between Cter of ZO-1 (1,619–1,748) and either DbpA (E–G) or occludin (H–J) for binding to ZPSG-1. (K and L) ZO-2 promotes ZO-1 interaction with DbpA. Immunoblot analysis of GST pull-downs using GST-DbpA as a bait, and full-length ZO-1 as a prey, in the presence of increasing amounts (μL of lysate indicated below each lane) of either vsv-tagged ZO-2 (K), or vsv-tagged p114-RhoGEF (L). Immunoblots (B, D, E, F, and H–M) show normalized loadings of preys (INPUT-prey, green), competing prey proteins (INPUT, blue), and GST pull-downs (preys in green) without or with competing proteins (blue). GST (negative control) and GST-fusion baits are in red, and their levels are shown in PonceauS-stained blots below immunoblots. Numbers indicate amino acid residues spanned in constructs. Preys were tagged either with GFP, HA, or vsv. FL-ZO-1 had no tag. Quantification of prey binding to GST-ZPSG- is shown in (B’) and (G). Binding of GFP-tagged ZPSG domains of ZO-1, ZO-2, and ZO-3 to either GST-DbpA or GST-occludin are shown in related Figure S5. Binding of Cter and DbpA to ZPSG-1 subdomains, competition experiments for binding to ZPSG-2, and determination of affinities of interaction of ZPSG-1 for either Cter or DbpA are shown in related Figure S6. Current Biology 2017 27, 3783-3795.e8DOI: (10.1016/j.cub.2017.11.014) Copyright © 2017 Elsevier Ltd Terms and Conditions

Figure 7 Force-Dependent Interaction of ZPSG and Cter In Vitro (A) Scheme of experimental setup. (B) Force-extension curves of rupturing/unfolding of ZPSG-FH1-Cter construct during multiple force-increase scans (1 pN/s). (C) Distributions of number of residues (top) and contour length (bottom) involved in the rupturing/unfolding events. (D) Force distributions for rupturing (>300 aa, top) and unfolding (<300 aa, bottom) of the ZPSG-Cter complex. (E) Typical force-extension curves of unfolding of ZPSG domain (loading rate of 1 pN/s). Left and right dashed boxes indicate regions where ZPSG/I27 domains unfolding occur, respectively. (F) Unfolding force distributions of ZPSG domain. N, number of events. Current Biology 2017 27, 3783-3795.e8DOI: (10.1016/j.cub.2017.11.014) Copyright © 2017 Elsevier Ltd Terms and Conditions