Faculty Opinions recommendation of Vaccinia virus-induced cell motility requires F11L-mediated inhibition of RhoA signaling.

Author(s):  
Ariel Blocker
Science ◽  
2006 ◽  
Vol 311 (5759) ◽  
pp. 377-381 ◽  
Author(s):  
Ferran Valderrama ◽  
João V. Cordeiro ◽  
Sibylle Schleich ◽  
Friedrich Frischknecht ◽  
Michael Way

RhoA signaling plays a critical role in many cellular processes, including cell migration. Here we show that the vaccinia F11L protein interacts directly with RhoA, inhibiting its signaling by blocking the interaction with its downstream effectors Rho-associated kinase (ROCK) and mDia. RNA interference–mediated depletion of F11L during infection resulted in an absence of vaccinia-induced cell motility and inhibition of viral morphogenesis. Disruption of the RhoA binding site in F11L, which resembles that of ROCK, led to an identical phenotype. Thus, inhibition of RhoA signaling is required for both vaccinia morphogenesis and virus-induced cell motility.


Cell Systems ◽  
2016 ◽  
Vol 2 (1) ◽  
pp. 38-48 ◽  
Author(s):  
Kate M. Byrne ◽  
Naser Monsefi ◽  
John C. Dawson ◽  
Andrea Degasperi ◽  
Jimi-Carlo Bukowski-Wills ◽  
...  

2018 ◽  
Vol 4 (2) ◽  
pp. 216-225 ◽  
Author(s):  
Corina Beerli ◽  
Artur Yakimovich ◽  
Samuel Kilcher ◽  
Glennys V. Reynoso ◽  
Gotthold Fläschner ◽  
...  

2013 ◽  
Vol 14 (1) ◽  
pp. 51-62 ◽  
Author(s):  
Yutaka Handa ◽  
Charlotte H. Durkin ◽  
Mark P. Dodding ◽  
Michael Way

Blood ◽  
2012 ◽  
Vol 119 (2) ◽  
pp. 513-520 ◽  
Author(s):  
Liat Nadav Dagan ◽  
Xiaoyu Jiang ◽  
Shruti Bhatt ◽  
Elena Cubedo ◽  
Klaus Rajewsky ◽  
...  

Abstract HGAL, a prognostic biomarker in patients with diffuse large B-cell lymphoma and classic Hodgkin lymphoma, inhibits lymphocyte and lymphoma cell motility by activating the RhoA signaling cascade and interacting with actin and myosin proteins. Although HGAL expression is limited to germinal center (GC) lymphocytes and GC-derived lymphomas, little is known about its regulation. miR-155 is implicated in control of GC reaction and lymphomagenesis. We demonstrate that miR-155 directly down-regulates HGAL expression by binding to its 3′-untranslated region, leading to decreased RhoA activation and increased spontaneous and chemoattractant-induced lymphoma cell motility. The effects of miR-155 on RhoA activation and cell motility can be rescued by transfection of HGAL lacking the miR-155 binding site. This inhibitory effect of miR-155 suggests that it may have a key role in the loss of HGAL expression on differentiation of human GC B cells to plasma cell. Furthermore, this effect may contribute to lymphoma cell dissemination and aggressiveness, characteristic of activated B cell–like diffuse large B-cell lymphoma typically expressing high levels of miR-155 and lacking HGAL expression.


Virology ◽  
2010 ◽  
Vol 404 (2) ◽  
pp. 231-239 ◽  
Author(s):  
Joachim Zwilling ◽  
Katja Sliva ◽  
Astrid Schwantes ◽  
Barbara Schnierle ◽  
Gerd Sutter

2017 ◽  
Vol 108 (3) ◽  
pp. 380-389 ◽  
Author(s):  
Miho Takaoka ◽  
Shun Ito ◽  
Yoshio Miki ◽  
Akira Nakanishi
Keyword(s):  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 681-681
Author(s):  
Xiaoyu Jiang ◽  
Xiaoqing Lu ◽  
Izidore S Lossos

Abstract Abstract 681 Expression of the Human Germinal center Associated Lymphoma (HGAL) gene is restricted to germinal center (GC) B-lymphocytes and GC-derived lymphomas. HGAL expression identifies lymphomas characterized by improved survival. Previously we have demonstrated that HGAL decreases spontaneous and chemoattractant-induced cell motility by 1) interaction with F-actin and myosin II, inhibiting the ability of myosin to translocate actin; 2) activating RhoA signaling by direct interactions with RhoA-specific guanine nucleotide exchange factors. We recently demonstrated that HGAL enhances B-cell receptor (BCR)-mediated Syk activation by directly binding to and enhancing Syk kinase activity. However, the precise molecular mechanisms of the HGAL effects on these signaling pathways and requirement for cofactors have yet to be described. Examination of the HGAL protein sequence revealed the presence of a putative growth factor receptor-bound protein 2 (Grb2) binding motif (YYENV; aa106-110). Grb2 is an abundant adaptor protein with important functions in multiple intracellular pathways, including BCR and RhoA signaling. We examined whether Grb2 may directly interact with the HGAL protein and be involved in regulation of HGAL-mediated signaling. Confocal microscopy studies demonstrated colocalization of HGAL and Grb2 proteins in Raji and HeLa cell lines, the latter transiently expressing HGAL protein. Concordantly, coimmunoprecipitation experiments in VAL and Raji lymphoma cell lines detected endogenous Grb2 in immunoprecipitates of endogenous HGAL and vice versa. GST pull down assay using recombinant GST-Grb2 failed to pull down non-phosphorylated TRX-HGAL protein. In contrast, TRX-HGAL protein in vitro phosphorylated by Lyn kinase was detected in the GST-Grb2 pull down, confirming a direct interaction between these proteins and demonstrating requirement for HGAL phosphorylation, as would be predicted from the YYENV binding motif. We further elucidated the relevance of this interaction by evaluating the effects of siRNA mediated knockdown of Grb2, HGAL or both proteins together on BCR and RhoA signaling in lymphoma cell lines. In accordance with our previous data, HGAL knockdown led to a significant decrease in phosphorylated Syk (Y352) following BCR stimulation. A similar decrease in Syk(P352) phosphorylation was observed following knockdown of Grb2 alone or simultaneously with HGAL. Concordantly, BCR-induced Ca2+ mobilization was decreased significantly and to a similar extent following HGAL or Grb2 knockdown. Concomitant knockdown of HGAL and Grb2 did not lead to additive or synergistic decrease in BCR-induced Ca2+ mobilization. Similarly, knockdown of HGAL or Grb2 decreased fibronectin(FN)-induced RhoA-GTP levels. To further elucidate the importance of the HGAL-Grb2 interaction, we have generated a plasmid encoding a HGAL(Y106FY107F) protein in which the putative Grb2 binding site was mutated. The HGAL(Y106FY107F) protein is not interacting with the Grb2 protein, as demonstrated by coimmunoprecipitation studies. Expression of the wild type HGAL protein in HeLa cells markedly increased both FN-induced RhoA-GTP levels and RhoA-mediated serum response factor (SRF)-induced transcriptional activity from the serum responsive element (SRE). In contrast, the HGAL(Y106FY107F) mutant did not affect either FN-induced RhoA-GTP levels or SRF-induced transcriptional activity from the SRE. These observations suggest that the HGAL-Grb2 interaction is important for mediating HGAL's effect on RhoA signaling pathway. HGAL was previously shown to inhibit lymphoma cell motility by activating the RhoA signaling pathway. Indeed, siRNA mediated HGAL knockdown significantly increased lymphoma cell chemotaxis in response to IL-6 and SDF1. Knockdown of Grb2 similarly increased IL-6-induced lymphoma chemotaxis, with no further increases in IL-6 induced chemotaxis observed flowing concomitant HGAL and Grb2 knockdown. In contrast, compared to the HGAL knockdown, knockdown of Grb2 significantly increased lymphoma cell chemotaxis in response to SDF-1. Concomitant knockdown of both Grb2 and HGAL resulted in an ever greater increase in lymphoma cell motility in response to SDF-1. In summary, our findings suggest that interaction of HGAL with Grb2 protein enhances both RhoA and B-cell receptor signaling and may be necessary for mediating at least some of the HGAL effects. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 33 (22) ◽  
pp. 4526-4537 ◽  
Author(s):  
Anne von Thun ◽  
Christian Preisinger ◽  
Oliver Rath ◽  
Juliane P. Schwarz ◽  
Chris Ward ◽  
...  

In certainRasmutant cell lines, the inhibition of extracellular signal-regulated kinase (ERK) signaling increases RhoA activity and inhibits cell motility, which was attributed to a decrease in Fra-1 levels. Here we report a Fra-1-independent augmentation of RhoA signaling during short-term inhibition of ERK signaling. Using mass spectrometry-based proteomics, we identified guanine exchange factor H1 (GEF-H1) as mediating this effect. ERK binds to the Rho exchange factor GEF-H1 and phosphorylates it on S959, causing inhibition of GEF-H1 activity and a consequent decrease in RhoA activity. Knockdown experiments and expression of a nonphosphorylatable S959A GEF-H1 mutant showed that this site is crucial in regulating cell motility and invasiveness. Thus, we identified GEF-H1 as a critical ERK effector that regulates motility, cell morphology, and invasiveness.


1998 ◽  
Vol 72 (12) ◽  
pp. 9924-9933 ◽  
Author(s):  
Christopher M. Sanderson ◽  
Geoffrey L. Smith

ABSTRACT Vaccinia virus (VV) induces two forms of cell motility: cell migration, which is dependent on the expression of early genes, and the formation of cellular projections, which requires the expression of late genes. The need for viral gene expression prior to cell motility suggests that VV proteins may affect how infected cells interact with the extracellular matrix. To address this, we have analyzed changes in cell-matrix adhesion after infection of BS-C-1 cells with VV. Whereas uninfected cells round up and detach from the culture flask in the presence of EGTA, infected cells remain attached to the culture flask with a stellate morphology. Ca2+-independent cell-matrix adhesion was evident by 10 h postinfection, after the onset of cell motility but before the formation of virus-induced cellular projections. Progression to Ca2+-independent adhesion required the expression of late viral genes but not the formation of intracellular enveloped virus particles or intracellular actin tails. Analyses of specific matrix proteins identified vitronectin and fibronectin as optimal ligands for Ca2+-independent adhesion and the formation of cellular projections. Adhesion to fibronectin was mediated via RGD motifs alone and was not inhibited by 500 μg of heparin/ml. Kistrin, a disintegrin which binds preferentially to the αvβ3 (vitronectin/fibronectin) receptor inhibited the formation of cellular projections without disrupting preformed matrix interactions. Finally, we show that Ca2+-independent cell-matrix adhesion is a dynamic process which mediates changes in the morphology of VV-infected cells and uninfected cells which exhibit a transformed phenotype.


Sign in / Sign up

Export Citation Format

Share Document