by Ines Brandherm, Jennifer Disse, Dagmar Zeuschner, and Volker Gerke

Slides:



Advertisements
Similar presentations
Volume 4, Issue 4, Pages (April 2015)
Advertisements

by Wei Zhang, and Robert W. Colman
Interaction between G Protein-Coupled Receptor 143 and Tyrosinase: Implications for Understanding Ocular Albinism Type 1  Elisabetta De Filippo, Anke.
Phospholipase D2 acts as an essential adaptor protein in the activation of Syk in antigen-stimulated mast cells by Jun Ho Lee, Young Mi Kim, Nam Wook Kim,
Patient-derived C-terminal mutation of FANCI causes protein mislocalization and reveals putative EDGE motif function in DNA repair by Luca Colnaghi, Mathew.
by Kesheng Dai, Richard Bodnar, Michael C. Berndt, and Xiaoping Du
The ER-Mitochondria Tethering Complex VAPB-PTPIP51 Regulates Autophagy
Federico Dajas-Bailador, Emma V. Jones, Alan J. Whitmarsh 
Volume 20, Issue 5, Pages (May 2011)
Lipopolysaccharide Activates Caspase-1 (Interleukin-1–Converting Enzyme) in Cultured Monocytic and Endothelial Cells by Ralf R. Schumann, Claus Belka,
Volume 11, Issue 4, Pages (April 2003)
IL-10 disrupts the Brd4-docking sites to inhibit LPS-induced CXCL8 and TNF-α expression in monocytes: Implications for chronic obstructive pulmonary disease 
by Xingming Deng, Fengqin Gao, and W. Stratford May
by Mi-Ae Kang, Su-Young Yun, and Jonghwa Won
Volume 57, Issue 3, Pages (February 2015)
Volume 5, Issue 5, Pages (November 2003)
Myeloma-derived Dickkopf-1 disrupts Wnt-regulated osteoprotegerin and RANKL production by osteoblasts: a potential mechanism underlying osteolytic bone.
LPS induces CD40 gene expression through the activation of NF-κB and STAT-1α in macrophages and microglia by Hongwei Qin, Cynthia A. Wilson, Sun Jung Lee,
Volume 11, Issue 6, Pages (June 2012)
by Thomas D. Nightingale, Krupa Pattni, Alistair N. Hume, Miguel C
Volume 22, Issue 5, Pages (May 2012)
Volume 62, Issue 4, Pages (October 2002)
Tie2-R849W Mutant in Venous Malformations Chronically Activates a Functional STAT1 to Modulate Gene Expression  Hsiao-Tang Hu, Yi-Hsien Huang, Yi-Ann.
DNA and factor VII–activating protease protect against the cytotoxicity of histones by Gerben Marsman, Helen von Richthofen, Ingrid Bulder, Florea Lupu,
Volume 27, Issue 1, Pages (July 2007)
Cyclin C/Cdk3 Promotes Rb-Dependent G0 Exit
MUC1 Oncoprotein Stabilizes and Activates Estrogen Receptor α
Robert L.S Perry, Maura H Parker, Michael A Rudnicki  Molecular Cell 
by Silvia Mele, Stephen Devereux, Andrea G
Volume 29, Issue 3, Pages (February 2008)
Arachidonic acid induces ERK activation via Src SH2 domain association with the epidermal growth factor receptor  L.D. Alexander, Y. Ding, S. Alagarsamy,
HRTV and SFTSV NSs, but not UUKV NSs, inhibits JAK/STAT IFN signaling.
Volume 22, Issue 15, Pages (August 2012)
Transcription Factor MIZ-1 Is Regulated via Microtubule Association
Phospholipid Scramblase 1 Mediates Type I Interferon-Induced Protection against Staphylococcal α-Toxin  Miroslaw Lizak, Timur O. Yarovinsky  Cell Host.
Jungmook Lyu, Vicky Yamamoto, Wange Lu  Developmental Cell 
A JNK-Dependent Pathway Is Required for TNFα-Induced Apoptosis
MUC1 Oncoprotein Stabilizes and Activates Estrogen Receptor α
Upregulation of Tenascin-C Expression by IL-13 in Human Dermal Fibroblasts via the Phosphoinositide 3-kinase/Akt and the Protein Kinase C Signaling Pathways 
Histamine Inhibits the Production of Interferon-induced Protein of 10 kDa in Human Squamous Cell Carcinoma and Melanoma  Naoko Kanda, Shinichi Watanabe 
Naoko Kanda, Shinichi Watanabe  Journal of Investigative Dermatology 
FOXO3a Is Activated in Response to Hypoxic Stress and Inhibits HIF1-Induced Apoptosis via Regulation of CITED2  Walbert J. Bakker, Isaac S. Harris, Tak.
Lizhong Xu, Veronica Lubkov, Laura J. Taylor, Dafna Bar-Sagi 
The Actin-Bundling Protein Palladin Is an Akt1-Specific Substrate that Regulates Breast Cancer Cell Migration  Y. Rebecca Chin, Alex Toker  Molecular.
Volume 8, Issue 5, Pages (November 2008)
PPARδ Is a Type 1 IFN Target Gene and Inhibits Apoptosis in T Cells
Volume 20, Issue 4, Pages (November 2005)
Temporal Regulation of Salmonella Virulence Effector Function by Proteasome- Dependent Protein Degradation  Tomoko Kubori, Jorge E. Galán  Cell  Volume.
Rsk1 mediates a MEK–MAP kinase cell survival signal
LRP4 Serves as a Coreceptor of Agrin
Volume 19, Issue 14, Pages (July 2009)
ASPP2 Regulates Epithelial Cell Polarity through the PAR Complex
Volume 25, Issue 5, Pages (March 2007)
Mst1 Is an Interacting Protein that Mediates PHLPPs' Induced Apoptosis
Volume 129, Issue 2, Pages (April 2007)
Volume 125, Issue 4, Pages (May 2006)
1α,25-Dihydroxyvitamin D3 Stimulates Activator Protein 1 DNA-Binding Activity by a Phosphatidylinositol 3-Kinase/Ras/MEK/Extracellular Signal Regulated.
Defective RAB1B-related megakaryocytic ER-to-Golgi transport in RUNX1 haplodeficiency: impact on von Willebrand factor by Gauthami Jalagadugula, Lawrence.
Growth Factor-Dependent Trafficking of Cerebellar NMDA Receptors via Protein Kinase B/Akt Phosphorylation of NR2C  Bo-Shiun Chen, Katherine W. Roche 
Naoko Kanda, Shinichi Watanabe  Journal of Investigative Dermatology 
IGF-II and IGFBP-5 rescue the atrophy of myotubes induced by adipocytes. IGF-II and IGFBP-5 rescue the atrophy of myotubes induced by adipocytes. A–D:
Volume 5, Issue 4, Pages (April 2007)
Paracrine Apoptotic Effect of p53 Mediated by Tumor Suppressor Par-4
Suman Paul, Anuj K. Kashyap, Wei Jia, You-Wen He, Brian C. Schaefer 
Volume 23, Issue 2, Pages (August 2005)
Phosphorylation of the pro-apoptotic protein BAD on serine 155, a novel site, contributes to cell survival  K. Virdee, P.A. Parone, A.M. Tolkovsky  Current.
Volume 65, Issue 5, Pages e4 (March 2017)
Cdk1 Negatively Regulates Midzone Localization of the Mitotic Kinesin Mklp2 and the Chromosomal Passenger Complex  Stefan Hümmer, Thomas U. Mayer  Current.
The adaptor protein Lad associates with the G protein β subunit and mediates chemokine-dependent T-cell migration by Dongsu Park, Inyoung Park, Deogwon.
The interaction between PARsylated BRCA1 and RAP80 is required for maintaining BRCA1–RAP80–PARP1 complex integrity after DNA damage and normal HRR regulation.
Presentation transcript:

by Ines Brandherm, Jennifer Disse, Dagmar Zeuschner, and Volker Gerke cAMP-induced secretion of endothelial von Willebrand factor is regulated by a phosphorylation/dephosphorylation switch in annexin A2 by Ines Brandherm, Jennifer Disse, Dagmar Zeuschner, and Volker Gerke Blood Volume 122(6):1042-1051 August 8, 2013 ©2013 by American Society of Hematology

Forskolin-evoked secretion of VWF is inhibited by knockdown of AnxA2 and S100A10. Forskolin-evoked secretion of VWF is inhibited by knockdown of AnxA2 and S100A10. HUVECs were transiently transfected with either nontargeting siRNA duplexes (ct) or duplexes specific for AnxA2 or S100A10. (A-B) Forty-eight hours after transfection, PNSs were prepared and analyzed by immunoblotting for the presence of AnxA2 and S100A10. Probing for α-tubulin and vimentin served as loading controls. Signal intensities were quantified as described in the supplemental Methods, and results from different experiments are summarized in the right panels (n = number of independent experiments). Statistical significance was evaluated by unpaired Student t test. Bars represent mean ± standard error of the mean (SEM). (C-D) Quantification of VWF secretion in response to forskolin from HUVECs depleted of (C) AnxA2 or (D) S100A10. Forty-eight hours following transfection with siRNA duplexes, HUVECs were subjected to agonist treatment of 20 minutes. The amount of VWF released into the cell culture supernatant was then determined as described in Methods. Effects on secretion were quantified in ≥3 independent experiments, and 1-way ANOVA with Bonferroni test was performed to evaluate statistical significance. Bars represent mean ± SEM. Ines Brandherm et al. Blood 2013;122:1042-1051 ©2013 by American Society of Hematology

Subcellular localization of Anx A2 and S100A10. Subcellular localization of Anx A2 and S100A10. (A) HUVECs were incubated in basal medium or forskolin/3-Isobutyl-1-methylxanthine (IBMX)-containing stimulation medium for 20 minutes and subsequently fixed and permeabilized using 0.2% Triton-X100 in 4% paraformaldehyde (PFA)/phosphate-buffered saline. Cells were then stained with anti-AnxA2 (green) and anti-S100A10 (red) antibodies. 4,6 diamidino-2-phenylindole (DAPI) was used to label the nuclei (blue). Bars represent 10 µm. (B) For the quantification of the subcellular localization of AnxA2 and S100A10, signal intensities of both proteins in the cytoplasm and at the plasma membrane of 10 single cells per experiment were analyzed using ImageJ software. Ratios of membrane vs cytosolic intensities were calculated in 3 independent experiments, and the unpaired Student t test was performed to evaluate statistical significance. Bars represent mean ± SEM. Ines Brandherm et al. Blood 2013;122:1042-1051 ©2013 by American Society of Hematology

The entire AnxA2-S100A10 complex is involved in cAMP-dependent VWF release from HUVECs. HUVECs were transfected with siRNA specific for AnxA2 or nontargeting siRNA (ct RNAi). The entire AnxA2-S100A10 complex is involved in cAMP-dependent VWF release from HUVECs. HUVECs were transfected with siRNA specific for AnxA2 or nontargeting siRNA (ct RNAi). (A) Forty-eight hours later, cells were subjected to a second transfection using control siRNA, AnxA2 siRNA, or AnxA2 siRNA plus an siRNA-insensitive AnxA2 expression vector (AII-Rr). PNSs were prepared and analyzed by immunoblotting using rabbit polyclonal anti-AnxA2 antibodies for the detection of the total (endogenous and ectopically expressed) AnxA2 (total) and mouse monoclonal anti-AnxA2 antibodies (clone H28) for the specific detection of only ectopically expressed AnxA2 (ect.; see supplemental Methods). Probing with monoclonal α-tubulin antibodies was included as a loading control. (B) HUVECs transiently transfected with AnxA2 siRNA plus rescue plasmid (AII-Rr) were stained with (left) anti-AnxA2 antibodies specifically labeling the ectopically expressed AnxA2 (clone H28) and (right) anti-S100A10 antibodies directly coupled to Alexa Fluor 594. Nuclei were labeled with DAPI (blue). Scale bars represent 10 µm. Note the efficient restoration of AnxA2 and endogenous S100A10 levels in cells expressing the AnxA2 rescue construct (AII-Rr). (C) Cells transfected with siRNA (AnxA2 or nontargeting siRNA) or siRNA plus rescue plasmid (AII-Rr) were subjected to secretagogue-free and forskolin/IBMX-containing stimulation media for 20 min. Subsequently, the amount of released VWF was analyzed by enzyme-linked immunosorbent assay (ELISA). The effect on secretion was quantified in sets of 13 independent experiments, and 1-way ANOVA with Bonferroni test was performed to evaluate statistical significance. Bars represent mean ± SEM. (D) Forty-eight hours after the first siRNA transfection, HUVECs were subjected to a second round of transfection using either AnxA2 siRNA, AnxA2 siRNA plus a ubiquitinylation-resistant S100A10 mutant construct (YFP-S100A10-CΔ6), or control siRNA. Following another 48 h, PNS fractions were prepared and subjected to immunoblotting using rabbit polyclonal anti-AnxA2 and mouse monoclonal anti-S100A10 antibodies. Probing with monoclonal α-tubulin antibodies served as an internal loading control. Note the stability of the S100A10 mutant in cells depleted of AnxA2. (E) Transfections were carried out as described in D, and the amount of VWF released in response to forskolin was quantified by ELISA in sets of 3 independent experiments. One-way ANOVA with Bonferroni test was used for the evaluation of statistical significance. Bars represent mean ± SEM. Ines Brandherm et al. Blood 2013;122:1042-1051 ©2013 by American Society of Hematology

Forskolin induces dephosphorylation of AnxA2. Forskolin induces dephosphorylation of AnxA2. (A) HUVECs were incubated in forskolin/IBMX-containing stimulation medium or basal medium containing DMSO as vehicle control for 30 minutes, and PNSs were subjected to immunoprecipitation (IP) using anti–phosphoserine-coupled agarose. The amount of AnxA2 present in the PNS and the immunocomplexes was quantified by immunoblotting using mouse monoclonal anti-AnxA2 antibodies (clone HH7). One representative example is shown on the left. For quantification, AnxA2 levels in PNS fractions (input) were normalized to the respective level of the Ig heavy chain signals (internal loading control) and calculated as percentage of protein precipitated from unstimulated cells (right). Statistical significance of the results obtained from 3 independent experiments was evaluated by unpaired Student t test. (B) Serum-starved HUVECs were pretreated for 5 hours with 50 nM FK-506 or DMSO as vehicle control at 37°C. Subsequently, cells were incubated in forskolin/IBMX-containing stimulation or basal medium for 30 minutes, and PNSs were subjected to anti–phospho-serine IP as described in A. PNS and IP fractions were probed for AnxA2, and a representative immunoblot of 2 independent experiments is shown. Numbers below the blot indicate AnxA2 levels in the IPs that were calculated as percent of protein precipitated from unstimulated cells as described in A. (C) HUVECs were preincubated in medium containing FK-506 or DMSO (vehicle control) and then subjected to forskolin/IBMX treatment as outlined in Methods. The amount of VWF released into the cell culture supernatant was analyzed by ELISA, and statistical significance of the results obtained from 11 independent experiments was evaluated by unpaired Student t test. Bars represent mean ± SEM. The basal secretion is not affected by FK-506 treatment, whereas the calcineurin inhibitor significantly reduces the forskolin-induced secretion. Ines Brandherm et al. Blood 2013;122:1042-1051 ©2013 by American Society of Hematology

Phosphorylation of Ser-11 of AnxA2 interferes with S100A10 complex formation in MDCK cells. Phosphorylation of Ser-11 of AnxA2 interferes with S100A10 complex formation in MDCK cells. Subcellular localization of different AnxA2 mutants in MDCK cells stably transfected with the tetracycline-regulated XM-AnxA2 construct that induces AnxA2/S100A10 aggregates. MDCK-tTA-XM cells were cultivated either in (A) tetracycline-containing medium or in (B-E) the absence of tetracycline to induce expression of the XM construct. Subsequently, cells were transfected with the respective GFP-tagged AnxA2 constructs ([A-B] WT, [C] S11A, [D] S11D) or (E) a GFP expression vector as control. Twenty-four hours after transfection, cells were fixed, permeabilized, and stained with anti-S100A10 antibodies (clone H21) that recognize the XM derivative. WT- and S11A-AnxA2 but not S11D-AnxA2 are recruited to these clusters. Scale bars represent 10 µm. (F) Colocalization coefficients of S100A10-stained XM clusters and the respective AnxA2-GFP constructs of 7 single cells per experiment were determined using BioImageXD software, and statistical significance of 3 independent experiments was analyzed by 1-way ANOVA with Bonferroni test. Bars represent mean ± SEM. Ines Brandherm et al. Blood 2013;122:1042-1051 ©2013 by American Society of Hematology

The phosphomimicking S11D mutation in AnxA2 attenuates the interaction between AnxA2 and S100A10 in HUVECs. HUVECs were transiently transfected with WT-, S11A-, or S11D-AnxA2, respectively. The phosphomimicking S11D mutation in AnxA2 attenuates the interaction between AnxA2 and S100A10 in HUVECs. HUVECs were transiently transfected with WT-, S11A-, or S11D-AnxA2, respectively. Forty-eight hours after transfection, PNSs were prepared and subjected to immunoprecipitation using mouse monoclonal antibodies only recognizing the ectopically expressed AnxA2 derivatives (clone H28; supplemental Methods). The amount of AnxA2 derivative (ect. AnxA2) and S100A10 present in the PNSs and the immunoprecipitates was quantified as described in supplemental Methods. For quantification, AnxA2 levels in IP fractions were normalized to the respective level in the PNS fraction (input), and the amount of coprecipitated S100A10 was calculated as percentage of precipitated AnxA2 (bar diagram). Statistical significance of the results obtained from 3 independent experiments was evaluated by 1-way ANOVA with the unpaired Student t test. Bars represent mean ± SEM. Ines Brandherm et al. Blood 2013;122:1042-1051 ©2013 by American Society of Hematology

AnxA2 containing a phosphomimicking mutation at Ser-11 does not promote forskolin-induced VWF secretion. AnxA2 containing a phosphomimicking mutation at Ser-11 does not promote forskolin-induced VWF secretion. Forty-eight hours after transfection with AnxA2 siRNA or nontargeting siRNA (ct RNAi), HUVECs were transfected a second time either with siRNA specific for AnxA2, control siRNA, or with AnxA2 siRNA together with siRNA-insensitive AnxA2 mutant constructs (S11A or S11D). Rescue of AnxA2 protein levels following expression of the (A) AII-Rr-S11A or (B) AII-Rr-S11D constructs was verified by immunoblotting using polyclonal rabbit anti-AnxA2 antibodies for the detection of endogenous and exogenous AnxA2 (total) or monoclonal mouse anti-AnxA2 antibodies (clone H28) for the detection of only ectopically expressed S11A- and S11D-AnxA2 (ect.). Probing with anti–α-tubulin antibodies was used as internal loading control. (C-D) HUVECs transiently transfected with AnxA2 siRNA plus the respective rescue plasmids ([C] AII-Rr-S11A and [D] AII-Rr-S11D) were stained with (left) anti-AnxA2 antibodies (clone H28 to label ectopically expressed AnxA2) and (right) anti-S100A10 antibodies directly coupled to Alexa Fluor 594. Nuclei were labeled with DAPI (blue). Note the restoration of endogenous S100A10 levels in cells ectopically expressing AII-Rr-S11A but not in cells expressing AII-Rr-S11D. Scale bars represent 10 µm. (E-F) HUVECs transfected with siRNA and the respective rescue constructs were subjected to forskolin/IBMX treatment, and VWF secretion was analyzed by ELISA. The effect on secretion was quantified in (C) 9 and (D) 11 independent experiments. One-way ANOVA with Bonferroni test was used for the quantification of statistical significance. Bars represent mean ± SEM. Ines Brandherm et al. Blood 2013;122:1042-1051 ©2013 by American Society of Hematology