@article{buglak_bougaran_kulikauskas_liu_monaghan-benson_gold_marvin_burciu_tanke_oatley_et al._2023, title={Nuclear SUN1 stabilizes endothelial cell junctions via microtubules to regulate blood vessel formation}, volume={12}, ISSN={["2050-084X"]}, DOI={10.7554/eLife.83652}, abstractNote={Endothelial cells line all blood vessels, where they coordinate blood vessel formation and the blood-tissue barrier via regulation of cell-cell junctions. The nucleus also regulates endothelial cell behaviors, but it is unclear how the nucleus contributes to endothelial cell activities at the cell periphery. Here, we show that the nuclear-localized linker of the nucleoskeleton and cytoskeleton (LINC) complex protein SUN1 regulates vascular sprouting and endothelial cell-cell junction morphology and function. Loss of murine endothelial Sun1 impaired blood vessel formation and destabilized junctions, angiogenic sprouts formed but retracted in SUN1-depleted sprouts, and zebrafish vessels lacking Sun1b had aberrant junctions and defective cell-cell connections. At the cellular level, SUN1 stabilized endothelial cell-cell junctions, promoted junction function, and regulated contractility. Mechanistically, SUN1 depletion altered cell behaviors via the cytoskeleton without changing transcriptional profiles. Reduced peripheral microtubule density, fewer junction contacts, and increased catastrophes accompanied SUN1 loss, and microtubule depolymerization phenocopied effects on junctions. Depletion of GEF-H1, a microtubule-regulated Rho activator, or the LINC complex protein nesprin-1 rescued defective junctions of SUN1-depleted endothelial cells. Thus, endothelial SUN1 regulates peripheral cell-cell junctions from the nucleus via LINC complex-based microtubule interactions that affect peripheral microtubule dynamics and Rho-regulated contractility, and this long-range regulation is important for proper blood vessel sprouting and junction integrity.}, journal={ELIFE}, author={Buglak, Danielle B. and Bougaran, Pauline and Kulikauskas, Molly R. and Liu, Ziqing and Monaghan-Benson, Elizabeth and Gold, Ariel L. and Marvin, Allison P. and Burciu, Andrew and Tanke, Natalie T. and Oatley, Morgan and et al.}, year={2023}, month={Mar} } @article{belaadi_pernet_aureille_chadeuf_rio_vaillant_vitiello_lafanechere_loirand_guilluy_2022, title={SUN2 regulates mitotic duration in response to extracellular matrix rigidity}, volume={119}, ISSN={["1091-6490"]}, DOI={10.1073/pnas.2116167119}, abstractNote={How cells adjust their growth to the spatial and mechanical constraints of their surrounding environment is central to many aspects of biology. Here, we examined how extracellular matrix (ECM) rigidity affects cell division. We found that cells divide more rapidly when cultured on rigid substrates. While we observed no effect of ECM rigidity on rounding or postmitotic spreading duration, we found that changes in matrix stiffness impact mitosis progression. We noticed that ECM elasticity up-regulates the expression of the linker of nucleoskeleton and cytoskeleton (LINC) complex component SUN2, which in turn promotes metaphase-to-anaphase transition by acting on mitotic spindle formation, whereas when cells adhere to soft ECM, low levels of SUN2 expression perturb astral microtubule organization and delay the onset of anaphase.}, number={45}, journal={PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA}, author={Belaadi, Nejma and Pernet, Lydia and Aureille, Julien and Chadeuf, Gilliane and Rio, Marc and Vaillant, Nathalie and Vitiello, Elisa and Lafanechere, Laurence and Loirand, Gervaise and Guilluy, Christophe}, year={2022}, month={Nov} } @article{duchalais_guilluy_nedellec_touvron_bessard_touchefeu_bossard_boudin_louarn_neunlist_et al._2018, title={Colorectal Cancer Cells Adhere to and Migrate Along the Neurons of the Enteric Nervous System}, volume={5}, ISSN={2352-345X}, url={http://dx.doi.org/10.1016/J.JCMGH.2017.10.002}, DOI={10.1016/J.JCMGH.2017.10.002}, abstractNote={Background & AimsIn several types of cancers, tumor cells invade adjacent tissues by migrating along the resident nerves of the tumor microenvironment. This process, called perineural invasion, typically occurs along extrinsic nerves, with Schwann cells providing physical guidance for the tumor cells. However, in the colorectal cancer microenvironment, the most abundant nervous structures belong to the nonmyelinated intrinsic enteric nervous system (ENS). In this study, we investigated whether colon cancer cells interact with the ENS.MethodsTumor epithelial cells (TECs) from human primary colon adenocarcinomas and cell lines were cocultured with primary cultures of ENS and cultures of human ENS plexus explants. By combining confocal and atomic force microscopy, as well as video microscopy, we assessed tumor cell adhesion and migration on the ENS. We identified the adhesion proteins involved using a proteomics approach based on biotin/streptavidin interaction, and their implication was confirmed further using selective blocking antibodies.ResultsTEC adhered preferentially and with stronger adhesion forces to enteric nervous structures than to mesenchymal cells. TEC adhesion to ENS involved direct interactions with enteric neurons. Enteric neuron removal from ENS cultures led to a significant decrease in tumor cell adhesion. TECs migrated significantly longer and further when adherent on ENS compared with on mesenchymal cells, and their trajectory faithfully followed ENS structures. Blocking N-cadherin and L1CAM decreased TEC migration along ENS structures.ConclusionsOur data show that the enteric neuronal network guides tumor cell migration, partly via L1CAM and N-cadherin. These results open a new avenue of research on the underlying mechanisms and consequences of perineural invasion in colorectal cancer. In several types of cancers, tumor cells invade adjacent tissues by migrating along the resident nerves of the tumor microenvironment. This process, called perineural invasion, typically occurs along extrinsic nerves, with Schwann cells providing physical guidance for the tumor cells. However, in the colorectal cancer microenvironment, the most abundant nervous structures belong to the nonmyelinated intrinsic enteric nervous system (ENS). In this study, we investigated whether colon cancer cells interact with the ENS. Tumor epithelial cells (TECs) from human primary colon adenocarcinomas and cell lines were cocultured with primary cultures of ENS and cultures of human ENS plexus explants. By combining confocal and atomic force microscopy, as well as video microscopy, we assessed tumor cell adhesion and migration on the ENS. We identified the adhesion proteins involved using a proteomics approach based on biotin/streptavidin interaction, and their implication was confirmed further using selective blocking antibodies. TEC adhered preferentially and with stronger adhesion forces to enteric nervous structures than to mesenchymal cells. TEC adhesion to ENS involved direct interactions with enteric neurons. Enteric neuron removal from ENS cultures led to a significant decrease in tumor cell adhesion. TECs migrated significantly longer and further when adherent on ENS compared with on mesenchymal cells, and their trajectory faithfully followed ENS structures. Blocking N-cadherin and L1CAM decreased TEC migration along ENS structures. Our data show that the enteric neuronal network guides tumor cell migration, partly via L1CAM and N-cadherin. These results open a new avenue of research on the underlying mechanisms and consequences of perineural invasion in colorectal cancer.}, number={1}, journal={Cellular and Molecular Gastroenterology and Hepatology}, publisher={Elsevier BV}, author={Duchalais, Emilie and Guilluy, Christophe and Nedellec, Steven and Touvron, Melissa and Bessard, Anne and Touchefeu, Yann and Bossard, Céline and Boudin, Hélène and Louarn, Guy and Neunlist, Michel and et al.}, year={2018}, pages={31–49} } @article{marchand_umana_pichol-thievend_salza_ricard-blum_monnot_guilluy_muller_germain_2017, title={Lysyl oxidase like-2 (LOXL2) regulates endothelial mechanotransduction and 3D vascular morphogenesis through scaffolding of basement membrane}, volume={9}, ISSN={1878-6480}, url={http://dx.doi.org/10.1016/S1878-6480(17)30407-X}, DOI={10.1016/S1878-6480(17)30407-X}, number={2}, journal={Archives of Cardiovascular Diseases Supplements}, publisher={Elsevier BV}, author={Marchand, M. and Umana, C. and Pichol-Thievend, C. and Salza, R. and Ricard-Blum, S. and Monnot, C. and Guilluy, C. and Muller, L. and Germain, S.}, year={2017}, month={Apr}, pages={164} } @article{aureille_belaadi_guilluy_2017, title={Mechanotransduction via the nuclear envelope: a distant reflection of the cell surface}, volume={44}, ISSN={0955-0674}, url={http://dx.doi.org/10.1016/J.CEB.2016.10.003}, DOI={10.1016/J.CEB.2016.10.003}, abstractNote={As the largest and stiffest organelle in the cell, the nucleus can be subjected to significant forces generated by the cytoskeleton to adjust its shape and position, and accommodate the cellular machinery during cell migration, differentiation or division. As it was anticipated, recent work showed that mechanosensitive mechanisms exist in the nucleus and regulate its structure and function in response to mechanical force. While the molecular mechanisms that mediate this response are only beginning to be elucidated, the nuclear envelope seems to play a central role in this process. Here, we review these nuclear mechanosensitive mechanisms and highlight their functional homology with those located at the cell surface. Additionally, we discuss how these nuclear envelope mechanisms function during adhesion and migration, and how they participate in cytoskeletal organization, via direct physical contact or signaling event regulation.}, journal={Current Opinion in Cell Biology}, publisher={Elsevier BV}, author={Aureille, Julien and Belaadi, Néjma and Guilluy, Christophe}, year={2017}, month={Feb}, pages={59–67} } @article{burridge_guilluy_2016, title={Focal adhesions, stress fibers and mechanical tension}, volume={343}, ISSN={0014-4827}, url={http://dx.doi.org/10.1016/J.YEXCR.2015.10.029}, DOI={10.1016/J.YEXCR.2015.10.029}, abstractNote={Stress fibers and focal adhesions are complex protein arrays that produce, transmit and sense mechanical tension. Evidence accumulated over many years led to the conclusion that mechanical tension generated within stress fibers contributes to the assembly of both stress fibers themselves and their associated focal adhesions. However, several lines of evidence have recently been presented against this model. Here we discuss the evidence for and against the role of mechanical tension in driving the assembly of these structures. We also consider how their assembly is influenced by the rigidity of the substratum to which cells are adhering. Finally, we discuss the recently identified connections between stress fibers and the nucleus, and the roles that these may play, both in cell migration and regulating nuclear function.}, number={1}, journal={Experimental Cell Research}, publisher={Elsevier BV}, author={Burridge, Keith and Guilluy, Christophe}, year={2016}, month={Apr}, pages={14–20} } @article{marjoram_guilluy_burridge_2016, title={Using magnets and magnetic beads to dissect signaling pathways activated by mechanical tension applied to cells}, volume={94}, ISSN={1046-2023}, url={http://dx.doi.org/10.1016/J.YMETH.2015.09.025}, DOI={10.1016/J.YMETH.2015.09.025}, abstractNote={Cellular tension has implications in normal biology and pathology. Membrane adhesion receptors serve as conduits for mechanotransduction that lead to cellular responses. Ligand-conjugated magnetic beads are a useful tool in the study of how cells sense and respond to tension. Here we detail methods for their use in applying tension to cells and strategies for analyzing the results. We demonstrate the methods by analyzing mechanotransduction through VE-cadherin on endothelial cells using both permanent magnets and magnetic tweezers.}, journal={Methods}, publisher={Elsevier BV}, author={Marjoram, R.J. and Guilluy, C. and Burridge, K.}, year={2016}, month={Feb}, pages={19–26} } @article{guilluy_burridge_2015, title={Nuclear mechanotransduction: Forcing the nucleus to respond}, volume={6}, ISSN={1949-1034 1949-1042}, url={http://dx.doi.org/10.1080/19491034.2014.1001705}, DOI={10.1080/19491034.2014.1001705}, abstractNote={Cell phenotype and fate are driven by the mechanical properties of their surrounding environment. Changes in matrix rigidity or application of force have been shown to impact profoundly cell behavior and phenotype, demonstrating that the molecular mechanisms which “sense” and transduce these signals into biochemical pathways are central in cell biology. In this commentary, we discuss recent evidence showing that mechanotransduction mechanisms occur in the nucleus, allowing dynamic regulation of the nucleoskeleton in response to mechanical stress. We will review this nucleoskeletal response and its impact on both nuclear structure and function.}, number={1}, journal={Nucleus}, publisher={Informa UK Limited}, author={Guilluy, Christophe and Burridge, Keith}, year={2015}, month={Jan}, pages={19–22} } @article{collins_osborne_guilluy_chen_o’brien_reader_burridge_superfine_tzima_2014, title={Haemodynamic and extracellular matrix cues regulate the mechanical phenotype and stiffness of aortic endothelial cells}, volume={5}, ISSN={2041-1723}, url={http://dx.doi.org/10.1038/NCOMMS4984}, DOI={10.1038/NCOMMS4984}, abstractNote={Endothelial cells (ECs) lining blood vessels express many mechanosensors, including platelet endothelial cell adhesion molecule-1 (PECAM-1), that convert mechanical force into biochemical signals. While it is accepted that mechanical stresses and the mechanical properties of ECs regulate vessel health, the relationship between force and biological response remains elusive. Here we show that ECs integrate mechanical forces and extracellular matrix (ECM) cues to modulate their own mechanical properties. We demonstrate that the ECM influences EC response to tension on PECAM-1. ECs adherent on collagen display divergent stiffening and focal adhesion growth compared with ECs on fibronectin. This is because of protein kinase A (PKA)-dependent serine phosphorylation and inactivation of RhoA. PKA signalling regulates focal adhesion dynamics and EC compliance in response to shear stress in vitro and in vivo. Our study identifies an ECM-specific, mechanosensitive signalling pathway that regulates EC compliance and may serve as an atheroprotective mechanism that maintains blood vessel integrity in vivo.}, number={1}, journal={Nature Communications}, publisher={Springer Science and Business Media LLC}, author={Collins, Caitlin and Osborne, Lukas D. and Guilluy, Christophe and Chen, Zhongming and O’Brien, E. Tim, III and Reader, John S. and Burridge, Keith and Superfine, Richard and Tzima, Ellie}, year={2014}, month={Jun} } @article{guilluy_osborne_van landeghem_sharek_superfine_garcia-mata_burridge_2014, title={Isolated nuclei adapt to force and reveal a mechanotransduction pathway in the nucleus}, volume={16}, ISSN={1465-7392 1476-4679}, url={http://dx.doi.org/10.1038/NCB2927}, DOI={10.1038/NCB2927}, abstractNote={Mechanical forces influence many aspects of cell behaviour. Forces are detected and transduced into biochemical signals by force-bearing molecular elements located at the cell surface, in adhesion complexes or in cytoskeletal structures. The nucleus is physically connected to the cell surface through the cytoskeleton and the linker of nucleoskeleton and cytoskeleton (LINC) complex, allowing rapid mechanical stress transmission from adhesions to the nucleus. Although it has been demonstrated that nuclei experience force, the direct effect of force on the nucleus is not known. Here we show that isolated nuclei are able to respond to force by adjusting their stiffness to resist the applied tension. Using magnetic tweezers, we found that applying force on nesprin-1 triggers nuclear stiffening that does not involve chromatin or nuclear actin, but requires an intact nuclear lamina and emerin, a protein of the inner nuclear membrane. Emerin becomes tyrosine phosphorylated in response to force and mediates the nuclear mechanical response to tension. Our results demonstrate that mechanotransduction is not restricted to cell surface receptors and adhesions but can occur in the nucleus.}, number={4}, journal={Nature Cell Biology}, publisher={Springer Science and Business Media LLC}, author={Guilluy, Christophe and Osborne, Lukas D. and Van Landeghem, Laurianne and Sharek, Lisa and Superfine, Richard and Garcia-Mata, Rafael and Burridge, Keith}, year={2014}, month={Mar}, pages={376–381} } @article{bays_peng_tolbert_guilluy_angell_pan_superfine_burridge_demali_2014, title={Vinculin phosphorylation differentially regulates mechanotransduction at cell–cell and cell–matrix adhesions}, volume={205}, ISSN={1540-8140 0021-9525}, url={http://dx.doi.org/10.1083/JCB.201309092}, DOI={10.1083/JCB.201309092}, abstractNote={Cells experience mechanical forces throughout their lifetimes. Vinculin is critical for transmitting these forces, yet how it achieves its distinct functions at cell–cell and cell–matrix adhesions remains unanswered. Here, we show vinculin is phosphorylated at Y822 in cell–cell, but not cell–matrix, adhesions. Phosphorylation at Y822 was elevated when forces were applied to E-cadherin and was required for vinculin to integrate into the cadherin complex. The mutation Y822F ablated these activities and prevented cells from stiffening in response to forces on E-cadherin. In contrast, Y822 phosphorylation was not required for vinculin functions in cell–matrix adhesions, including integrin-induced cell stiffening. Finally, forces applied to E-cadherin activated Abelson (Abl) tyrosine kinase to phosphorylate vinculin; Abl inhibition mimicked the loss of vinculin phosphorylation. These data reveal an unexpected regulatory mechanism in which vinculin Y822 phosphorylation determines whether cadherins transmit force and provides a paradigm for how a shared component of adhesions can produce biologically distinct functions.}, number={2}, journal={The Journal of Cell Biology}, publisher={Rockefeller University Press}, author={Bays, Jennifer L. and Peng, Xiao and Tolbert, Catlin E. and Guilluy, Christophe and Angell, Ashley E. and Pan, Yuan and Superfine, Richard and Burridge, Keith and DeMali, Kris A.}, year={2014}, month={Apr}, pages={251–263} } @article{thompson_guilluy_xie_sen_brobst_yen_uzer_styner_case_burridge_et al._2013, title={Mechanically activated fyn utilizes mTORC2 to regulate RhoA and adipogenesis in mesenchymal stem cells}, volume={31}, ISSN={1066-5099}, url={http://dx.doi.org/10.1002/STEM.1476}, DOI={10.1002/STEM.1476}, abstractNote={Abstract}, number={11}, journal={STEM CELLS}, publisher={Wiley}, author={Thompson, William R. and Guilluy, Christophe and Xie, Zhihui and Sen, Buer and Brobst, Kaitlyn E. and Yen, Sherwin S. and Uzer, Gunes and Styner, Maya and Case, Natasha and Burridge, Keith and et al.}, year={2013}, month={Nov}, pages={2528–2537} } @article{lessey_guilluy_burridge_2012, title={From Mechanical Force to RhoA Activation}, volume={51}, ISSN={0006-2960 1520-4995}, url={http://dx.doi.org/10.1021/bi300758e}, DOI={10.1021/bi300758e}, abstractNote={Throughout their lives, all cells constantly experience and respond to various mechanical forces. These frequently originate externally but can also arise internally as a result of the contractile actin cytoskeleton. Mechanical forces trigger multiple signaling pathways. Several converge and result in the activation of the GTPase RhoA. In this review, we focus on the pathways by which mechanical force leads to RhoA regulation, especially when force is transmitted via cell adhesion molecules that mediate either cell-matrix or cell-cell interactions. We discuss both the upstream signaling events that lead to activation of RhoA and the downstream consequences of this pathway. These include not only cytoskeletal reorganization and, in a positive feedback loop, increased myosin-generated contraction but also profound effects on gene expression and differentiation.}, number={38}, journal={Biochemistry}, publisher={American Chemical Society (ACS)}, author={Lessey, Elizabeth C. and Guilluy, Christophe and Burridge, Keith}, year={2012}, month={Sep}, pages={7420–7432} } @article{collins_guilluy_welch_o’brien_hahn_superfine_burridge_tzima_2012, title={Localized Tensional Forces on PECAM-1 Elicit a Global Mechanotransduction Response via the Integrin-RhoA Pathway}, volume={22}, ISSN={0960-9822}, url={http://dx.doi.org/10.1016/j.cub.2012.08.051}, DOI={10.1016/j.cub.2012.08.051}, abstractNote={

Summary

Background

Mechanical forces regulate cell behavior and function during development, differentiation, and tissue morphogenesis. In the vascular system, forces produced by blood flow are critical determinants not only of morphogenesis and function, but also of pathological states such as atherosclerosis. Endothelial cells (ECs) have numerous mechanotransducers, including platelet endothelial cell adhesion molecule-1 (PECAM-1) at cell-cell junctions and integrins at cell-matrix adhesions. However, the processes by which forces are transduced to biochemical signals and subsequently translated into downstream effects are poorly understood.

Results

Here, we examine mechanochemical signaling in response to direct force application on PECAM-1. We demonstrate that localized tensional forces on PECAM-1 result in, surprisingly, global signaling responses. Specifically, force-dependent activation of phosphatidylinositol 3-kinase (PI3K) downstream of PECAM-1 promotes cell-wide activation of integrins and the small GTPase RhoA. These signaling events facilitate changes in cytoskeletal architecture, including growth of focal adhesions and adaptive cytoskeletal stiffening.

Conclusions

Taken together, our work provides the first evidence of a global signaling event in response to a localized mechanical stress. In addition, these data provide a possible mechanism for the differential stiffness of vessels exposed to distinct hemodynamic force patterns in vivo.}, number={22}, journal={Current Biology}, publisher={Elsevier BV}, author={Collins, Caitlin and Guilluy, Christophe and Welch, Christopher and O’Brien, E. Timothy and Hahn, Klaus and Superfine, Richard and Burridge, Keith and Tzima, Ellie}, year={2012}, month={Nov}, pages={2087–2094} } @article{guilluy_dubash_garcía-mata_2011, title={Analysis of RhoA and Rho GEF activity in whole cells and the cell nucleus}, volume={6}, ISSN={1754-2189 1750-2799}, url={http://dx.doi.org/10.1038/nprot.2011.411}, DOI={10.1038/nprot.2011.411}, abstractNote={We have recently shown that a fraction of the total cellular pool of the small GTPase RhoA resides in the nucleus, and that the nuclear guanine nucleotide exchange factor (GEF) Net1 has a role in the regulation of its activity. In this protocol, we describe a method to measure both the activities of the nuclear pools of RhoA and Rho GEFs. This process required the development of a nuclear isolation protocol that is both fast and virtually free of cytosolic and membrane contaminants, as well as a redesign of existing RhoA and Rho GEF activity assays so that they work in nuclear samples. This protocol can be also used for other Rho GTPases and Rho GEFs, which have also been found in the nucleus. Completion of the procedure, including nuclear isolation and RhoA or Rho GEF activity assay, takes 1 h 40 min. We also include details of how to perform a basic assay of whole-cell extracts.}, number={12}, journal={Nature Protocols}, publisher={Springer Science and Business Media LLC}, author={Guilluy, Christophe and Dubash, Adi D and García-Mata, Rafael}, year={2011}, month={Dec}, pages={2050–2060} } @article{sen_guilluy_xie_case_styner_thomas_oguz_rubin_burridge_rubin_2011, title={Mechanically Induced Focal Adhesion Assembly Amplifies Anti-Adipogenic Pathways in Mesenchymal Stem Cells}, volume={29}, ISSN={1066-5099}, url={http://dx.doi.org/10.1002/stem.732}, DOI={10.1002/stem.732}, abstractNote={Abstract}, number={11}, journal={STEM CELLS}, publisher={Wiley}, author={Sen, Buer and Guilluy, Christophe and Xie, Zhihui and Case, Natasha and Styner, Maya and Thomas, Jacob and Oguz, Ipek and Rubin, Clinton and Burridge, Keith and Rubin, Janet}, year={2011}, month={Oct}, pages={1829–1836} } @article{guilluy_garcia-mata_burridge_2011, title={Rho protein crosstalk: another social network?}, volume={21}, ISSN={0962-8924}, url={http://dx.doi.org/10.1016/j.tcb.2011.08.002}, DOI={10.1016/j.tcb.2011.08.002}, abstractNote={Many fundamental processes in cell biology are regulated by Rho GTPases, including cell adhesion, migration and differentiation. While regulating cellular functions, members of the Rho protein family cooperate or antagonize each other. The resulting molecular network exhibits many levels of interaction dynamically regulated in time and space. In the first part of this review we describe the main mechanisms of this crosstalk, which can occur at three different levels of the pathway: (i) through regulation of activity, (ii) through regulation of protein expression and stability, and (iii) through regulation of downstream signaling pathways. In the second part we illustrate the importance of Rho protein crosstalk with two examples: integrin-based adhesion and cell migration.}, number={12}, journal={Trends in Cell Biology}, publisher={Elsevier BV}, author={Guilluy, Christophe and Garcia-Mata, Rafael and Burridge, Keith}, year={2011}, month={Dec}, pages={718–726} } @article{guilluy_swaminathan_garcia-mata_timothy o’brien_superfine_burridge_2011, title={The Rho GEFs LARG and GEF-H1 regulate the mechanical response to force on integrins}, volume={13}, ISSN={1465-7392 1476-4679}, url={http://dx.doi.org/10.1038/ncb2254}, DOI={10.1038/ncb2254}, abstractNote={How individual cells respond to mechanical forces is of considerable interest to biologists as force affects many aspects of cell behaviour. The application of force on integrins triggers cytoskeletal rearrangements and growth of the associated adhesion complex, resulting in increased cellular stiffness, also known as reinforcement. Although RhoA has been shown to play a role during reinforcement, the molecular mechanisms that regulate its activity are unknown. By combining biochemical and biophysical approaches, we identified two guanine nucleotide exchange factors (GEFs), LARG and GEF-H1, as key molecules that regulate the cellular adaptation to force. We show that stimulation of integrins with tensional force triggers activation of these two GEFs and their recruitment to adhesion complexes. Surprisingly, activation of LARG and GEF-H1 involves distinct signalling pathways. Our results reveal that LARG is activated by the Src family tyrosine kinase Fyn, whereas GEF-H1 catalytic activity is enhanced by ERK downstream of a signalling cascade that includes FAK and Ras.}, number={6}, journal={Nature Cell Biology}, publisher={Springer Science and Business Media LLC}, author={Guilluy, Christophe and Swaminathan, Vinay and Garcia-Mata, Rafael and Timothy O’Brien, E. and Superfine, Richard and Burridge, Keith}, year={2011}, month={May}, pages={722–727} } @article{shen_tolbert_guilluy_swaminathan_berginski_burridge_superfine_campbell_2011, title={The Vinculin C-terminal Hairpin Mediates F-actin Bundle Formation, Focal Adhesion, and Cell Mechanical Properties}, volume={286}, ISSN={0021-9258 1083-351X}, url={http://dx.doi.org/10.1074/jbc.M111.244293}, DOI={10.1074/jbc.M111.244293}, abstractNote={Vinculin is an essential and highly conserved cell adhesion protein, found at both focal adhesions and adherens junctions, where it couples integrins or cadherins to the actin cytoskeleton. Vinculin is involved in controlling cell shape, motility, and cell survival, and has more recently been shown to play a role in force transduction. The tail domain of vinculin (Vt) contains determinants necessary for binding and bundling of actin filaments. Actin binding to Vt has been proposed to induce formation of a Vt dimer that is necessary for cross-linking actin filaments. Results from this study provide additional support for actin-induced Vt self-association. Moreover, the actin-induced Vt dimer appears distinct from the dimer formed in the absence of actin. To better characterize the role of the Vt strap and carboxyl terminus (CT) in actin binding, Vt self-association, and actin bundling, we employed smaller amino-terminal (NT) and CT deletions that do not perturb the structural integrity of Vt. Although both NT and CT deletions retain actin binding, removal of the CT hairpin (1061–1066) selectively impairs actin bundling in vitro. Moreover, expression of vinculin lacking the CT hairpin in vinculin knock-out murine embryonic fibroblasts affects the number of focal adhesions formed, cell spreading as well as cellular stiffening in response to mechanical force.}, number={52}, journal={Journal of Biological Chemistry}, publisher={American Society for Biochemistry & Molecular Biology (ASBMB)}, author={Shen, Kai and Tolbert, Caitlin E. and Guilluy, Christophe and Swaminathan, Vinay S. and Berginski, Matthew E. and Burridge, Keith and Superfine, Richard and Campbell, Sharon L.}, year={2011}, month={Nov}, pages={45103–45115} } @article{boulter_garcia-mata_guilluy_dubash_rossi_brennwald_burridge_2010, title={Regulation of Rho GTPase crosstalk, degradation and activity by RhoGDI1}, volume={12}, ISSN={1465-7392 1476-4679}, url={http://dx.doi.org/10.1038/ncb2049}, DOI={10.1038/ncb2049}, abstractNote={At steady state, most Rho GTPases are bound in the cytosol to Rho guanine nucleotide dissociation inhibitors (RhoGDIs). RhoGDIs have generally been considered to hold Rho proteins passively in an inactive state within the cytoplasm. Here we describe an evolutionarily conserved mechanism by which RhoGDI1 controls the homeostasis of Rho proteins in eukaryotic cells. We found that depletion of RhoGDI1 promotes misfolding and degradation of the cytosolic geranylgeranylated pool of Rho GTPases while activating the remaining membrane-bound fraction. Because RhoGDI1 levels are limiting, and Rho proteins compete for binding to RhoGDI1, overexpression of an exogenous Rho GTPase displaces endogenous Rho proteins bound to RhoGDI1, inducing their degradation and inactivation. These results raise important questions about the conclusions drawn from studies that manipulate Rho protein levels. In many cases the response observed may arise not simply from the overexpression itself but from additional effects on the levels and activity of other Rho GTPases as a result of competition for binding to RhoGDI1; this may require a re-evaluation of previously published studies that rely exclusively on these techniques.}, number={5}, journal={Nature Cell Biology}, publisher={Springer Science and Business Media LLC}, author={Boulter, Etienne and Garcia-Mata, Rafael and Guilluy, Christophe and Dubash, Adi and Rossi, Guendalina and Brennwald, Patrick J. and Burridge, Keith}, year={2010}, month={Apr}, pages={477–483} } @article{guilluy_brégeon_toumaniantz_rolli-derkinderen_retailleau_loufrani_henrion_scalbert_bril_torres_et al._2010, title={The Rho exchange factor Arhgef1 mediates the effects of angiotensin II on vascular tone and blood pressure}, volume={16}, ISSN={1078-8956 1546-170X}, url={http://dx.doi.org/10.1038/nm.2079}, DOI={10.1038/nm.2079}, abstractNote={Hypertension is one of the most frequent pathologies in the industrialized world. Although recognized to be dependent on a combination of genetic and environmental factors, its molecular basis remains elusive. Increased activity of the monomeric G protein RhoA in arteries is a common feature of hypertension. However, how RhoA is activated and whether it has a causative role in hypertension remains unclear. Here we provide evidence that Arhgef1 is the RhoA guanine exchange factor specifically responsible for angiotensin II-induced activation of RhoA signaling in arterial smooth muscle cells. We found that angiotensin II activates Arhgef1 through a previously undescribed mechanism in which Jak2 phosphorylates Tyr738 of Arhgef1. Arhgef1 inactivation in smooth muscle induced resistance to angiotensin II-dependent hypertension in mice, but did not affect normal blood pressure regulation. Our results show that control of RhoA signaling through Arhgef1 is central to the development of angiotensin II-dependent hypertension and identify Arhgef1 as a potential target for the treatment of hypertension.}, number={2}, journal={Nature Medicine}, publisher={Springer Science and Business Media LLC}, author={Guilluy, Christophe and Brégeon, Jérémy and Toumaniantz, Gilles and Rolli-Derkinderen, Malvyne and Retailleau, Kevin and Loufrani, Laurent and Henrion, Daniel and Scalbert, Elizabeth and Bril, Antoine and Torres, Raul M and et al.}, year={2010}, month={Jan}, pages={183–190} } @article{agard_guilluy_eddahibi_guignabert_izikki_tu_savale_humbert_fadel_adnot_et al._2009, title={Activation de Rho kinase par sérotonylation intracellulaire de la petite protéine G RhoA dans l’hypertension artérielle pulmonaire}, volume={30}, ISSN={0248-8663}, url={http://dx.doi.org/10.1016/j.revmed.2009.10.132}, DOI={10.1016/j.revmed.2009.10.132}, abstractNote={L'endothélium vasculaire pulmonaire synthétise de nombreuses substances vasoactives qui peuvent être schématiquement classées en facteurs vasodilatateurs et vasoconstricteurs. Parmi les premiers, le monoxyde d'azote (NO) et le facteur hyperpolarisant dérivé de l'endothélium (endothelial-derived hyperpolarizing factor, EDHF) ont été les plus étudiés alors que l'endothéline est probablement le plus paissant vasoconstricteur endogène connu jusqu'à ce jour. Le NO est un composé radicalaire inactivable par l'hémoglobine dont les caractéristiques physiques sont semblables à celles d'un gaz et les propriétés thérapeutiques assimilables à celles d'un dérivé nitré endogène. De ce fait, l'inhalation du gaz NO représente à l'heure actvelle l'un des traitements les plus prometteurs de l'hypertension artérielle pulmonaire persistance du nouveau-né. L'EDHF agit en activant les canaux potassiques dépendants de l'ATP. Sa nature chimique et son rôle physiologique restent mal connus. L'endothéline possède des propriétés complexes. Entraînant une puissante vasoconstriction lorsqu'elle est mise en présence des cellules musculaires lisses, elle peut également induire la libération du NO et de l'EDHF par la cellule endothéliale et entraîner un effect vasodilatateur indirect, médié par l'endothélium. Les voles de transduction relayant les effects cellulaires de l'endothéline different dans les deux cas: l'activation des récepteurs endothéliaux ET-B est à l'origine de la vasodilatation alors que l'activation des récepteurs ET-A du muscle lisse vasculaire est responsable de la vasoconstriction. Cetle diversité des mécanismes cellulaires souligne l'intérêt d'une incilleure compréhension de la physiologie cellulaire relative à la circulation pulmonaire, afin de mieux cibler la cellule sur laquelle on pourrait agir pour corriger l'anomalia fonctionnelle responsable des dérèglements organiques observées en clinique.Pulmonary vascular endothelium synthesizes and releases two major groups of vasoactive substances, namely the endothelium-derived relaxing and contracting factors. Among the former, the effects of nitric oxide (NO), formely known as endothelium-derived relaxing factor (EDRF), and those of the so-called endothelium-derived hyperpolarizing factor (EDHF) have been extensively investigated. Among the latter, endothelin is probably one of the most potent endogenous vasoconstrictors. NO is a free radical which can be readily inactivated by hemoglobin. NO has all the characteristics of a gas, whereas its pharmacological properties are consistent with those of an endogenous nitrovasodilator. Therefore, inhalation of the gas NO is now considered as one of the most promising means to treat persistent pulmonary hypertension of the newborn. EDHF relaxes vascular smooth muscle through activation of ATP-dependent potassium channels. Both the chemical nature and the physiological role of EDHF are still unclear. The pharmacological properties of endothelin are far from being unequivocal. It is a potent vasoconstrictor when it directly acts on vascular smooth muscle. However, it can also induce the release of NO and EDHF, hence causing vasorelaxation. These effects of endothelin are mediated by various transduction pathways. Activations of ET-B receptors located on endothelium on the one hand, and ET-A receptors located on smooth muscle on the other hand, are responsible for relaxation and constriction of vascular smooth muscle, respectively. Such highly complex cellular mechanisms highlight the need for further insight into the physiology of the cell related to the palmonary circulation. This, in turn, will help to better define the target upon which one can try to correct the abnormal function of the cell underlying the pathophysiological processes}, journal={La Revue de Médecine Interne}, publisher={Elsevier BV}, author={Agard, C. and Guilluy, C. and Eddahibi, S. and Guignabert, C. and Izikki, M. and Tu, L. and Savale, L. and Humbert, M. and Fadel, E. and Adnot, S. and et al.}, year={2009}, month={Dec}, pages={S373} } @article{bregeon_guilluy_toumaniantz_rolliderkinderen_retailleau_loufrani_henrion_scalbert_bril_pacaud_et al._2009, title={I025 Smooth muscle specific deletion of the RhoA exchange factor Arhgef1 protects against Ang II-dependent hypertension}, volume={102}, ISSN={1875-2136}, url={http://dx.doi.org/10.1016/S1875-2136(09)72359-9}, DOI={10.1016/S1875-2136(09)72359-9}, abstractNote={Hypertension is one of the most frequent pathology in the industrialized world. Although it is recognized to be dependent on a combination of genetic and environmental factors, its molecular basis remains elusive. A possible candidate is the monomeric G protein RhoA which activates Rho kinase. However, how it is activated and whether it has a causative role in hypertension is still unknown. By in vitro experiments, we provide evidence that Arhgef1 is the RhoA guanine exchange factor specifically responsible for Angiotensin II (Ang II)-induced RhoA/Rho kinase activation in arterial smooth muscle cells. To analyze in vivo the role of Arhgef1, we generated mice lacking Arhgef1 specifically in smooth muscle cells (SM-Arhgef1-KO). We used Arhgef1lox/lox mice, which were mated to SMMHC-CreERT2 to produce SMMHC-CreERT2 ; Arhgef1lox/lox mice (SM-Arhgef1lox/lox). SM-Arhgef1-KO mice were then obtained by treating SM-Arhgef1lox/lox mice with tamoxifen. Remarkably, SM-Arhgef1-KO mice were resistant to hypertension induced by chronic Ang II infusion (sub-cutaneous osmotic pump, 1 μg/kg/min). Furthermore, Arhgef1 deletion after induction of hypertension by Ang II restored normal arterial pressure level. Basal arterial pressure and responses to vasoconstrictors other than Ang II (phenylephrine, U46619, endotheline) were not modified in SM-Arhgef1-KO mice. Our results show that Arhgef1 activation and its downstream RhoA/Rho kinase signaling are central to the development of Ang IIdependent hypertension. We identify Arhgef1 as an alternative potential target for the treatment of hypertension.}, journal={Archives of Cardiovascular Diseases}, publisher={Elsevier BV}, author={Bregeon, J. and Guilluy, C. and Toumaniantz, G. and Rolliderkinderen, M. and Retailleau, K. and Loufrani, L. and Henrion, D. and Scalbert, E. and Bril, A. and Pacaud, P. and et al.}, year={2009}, month={Mar}, pages={S95–S96} } @article{goueffic_guilluy_guerin_patra_pacaud_loirand_2006, title={Hyaluronan induces vascular smooth muscle cell migration through RHAMM-mediated PI3K-dependent Rac activation}, volume={72}, ISSN={0008-6363}, url={http://dx.doi.org/10.1016/J.CARDIORES.2006.07.017}, DOI={10.1016/J.CARDIORES.2006.07.017}, abstractNote={Hyaluronan (HA) is an important constituent of the extracellular matrix and is known to regulate cellular events through binding to CD44 and the receptor for HA-mediated motility (RHAMM). Here we investigated the role of these receptors and the signaling pathways involved in HA-mediated effects in arterial smooth muscle cells (ASMC).Effects of high-molecular weight HA (1 to 5 mg/ml) were analyzed in cultured ASMC from rat aorta.HA promoted actin stress fiber and lamellipodia formation and dose-dependently induced ASMC migration without effect on proliferation. Pull-down assay of Rho protein activity indicated that HA activated RhoA and Rac. HA-induced ASMC migration was not affected by the RhoA inhibitor Tat-C3 (10 microg/ml), the Rho kinase inhibitor Y-27632 (10 microM) and blocking anti-CD44 antibody ,but was reduced by the non-selective Rho protein inhibitor simvastatin (10 microM), the Rac inhibitor LT-toxin (1 mug/ml), small interfering RNA (siRNA) targeting Rac and the phosphatidyl inositol 3-kinase (PI3K) inhibitor LY294002 (25 microM), which also blocked HA-induced Rac activation. CD44 knockdown by siRNA inhibited HA-mediated RhoA activation without effect on ASMC migration. In contrast, siRNA targeting RHAMM inhibited both HA-induced migration and Rac activation.High-molecular weight HA independently activates RhoA and Rac through CD44 and RHAMM, respectively. HA-induced migration depends exclusively on RHAMM-mediated PI3K-dependent Rac activation.}, number={2}, journal={Cardiovascular Research}, publisher={Oxford University Press (OUP)}, author={Goueffic, Y and Guilluy, C and Guerin, P and Patra, P and Pacaud, P and Loirand, G}, year={2006}, month={Nov}, pages={339–348} } @article{loirand_guilluy_pacaud_2006, title={Regulation of Rho Proteins by Phosphorylation in the Cardiovascular System}, volume={16}, ISSN={1050-1738}, url={http://dx.doi.org/10.1016/j.tcm.2006.03.010}, DOI={10.1016/j.tcm.2006.03.010}, abstractNote={The small G protein Rho signaling pathways are recognized as major regulators of cardiovascular functions, and activation of Rho proteins appears to be a common component for the pathogenesis of hypertension and vascular proliferative disorders. Rho proteins are tightly regulated, and recent evidence suggests that modulation of Rho protein signaling by phosphorylation of Rho proteins provides an additional simple mechanism for coordinating Rho protein functions. This regulation by phosphorylation is particularly important in the arterial wall, where RhoA protein expressed in vascular smooth muscle cells is controlled by the endothelium through the nitric oxide/cGMP-dependent kinase pathway.}, number={6}, journal={Trends in Cardiovascular Medicine}, publisher={Elsevier BV}, author={Loirand, Gervaise and Guilluy, Christophe and Pacaud, Pierre}, year={2006}, month={Aug}, pages={199–204} } @article{guilluy_rolli-derkinderen_tharaux_melino_pacaud_loirand_2006, title={Transglutaminase-dependent RhoA Activation and Depletion by Serotonin in Vascular Smooth Muscle Cells}, volume={282}, ISSN={0021-9258 1083-351X}, url={http://dx.doi.org/10.1074/jbc.M604195200}, DOI={10.1074/jbc.M604195200}, abstractNote={The small G protein RhoA plays a major role in several vascular processes and cardiovascular disorders. Here we analyze the mechanisms of RhoA regulation by serotonin (5-HT) in arterial smooth muscle. 5-HT (0.1–10 μm) induced activation of RhoA followed by RhoA depletion at 24–72 h. Inhibition of 5-HT1 receptors reduced the early phase of RhoA activation but had no effect on 5-HT-induced delayed RhoA activation and depletion, which were suppressed by the 5-HT transporter inhibitor fluoxetine and the transglutaminase inhibitor monodansylcadaverin and in type 2 transglutaminase-deficient smooth muscle cells. Coimmunoprecipitations demonstrated that 5-HT associated with RhoA both in vitro and in vivo. This association was calcium-dependent and inhibited by fluoxetine and monodansylcadaverin. 5-HT promotes the association of RhoA with the E3 ubiquitin ligase Smurf1, and 5-HT-induced RhoA depletion was inhibited by the proteasome inhibitor MG132 and the RhoA inhibitor Tat-C3. Simvastatin, the Rho kinase inhibitor Y-27632, small interfering RNA-mediated RhoA gene silencing, and long-term 5-HT stimulation induced Akt activation. In contrast, inhibition of 5-HT-mediated RhoA degradation by MG132 prevented 5-HT-induced Akt activation. Long-term 5-HT stimulation also led to the inhibition of the RhoA/Rho kinase component of arterial contraction. Our data provide evidence that 5-HT, internalized through the 5-HT transporter, is transamidated to RhoA by transglutaminase. Transamidation of RhoA leads to RhoA activation and enhanced proteasomal degradation, which in turn is responsible for Akt activation and contraction inhibition. The observation of transamidation of 5-HT to RhoA in pulmonary artery of hypoxic rats suggests that this process could participate in pulmonary artery remodeling and hypertension.}, number={5}, journal={Journal of Biological Chemistry}, publisher={American Society for Biochemistry & Molecular Biology (ASBMB)}, author={Guilluy, Christophe and Rolli-Derkinderen, Malvyne and Tharaux, Pierre-Louis and Melino, Gerry and Pacaud, Pierre and Loirand, Gervaise}, year={2006}, month={Dec}, pages={2918–2928} } @article{guilluy_sauzeau_rolli-derkinderen_guérin_sagan_pacaud_loirand_2005, title={Inhibition of RhoA/Rho kinase pathway is involved in the beneficial effect of sildenafil on pulmonary hypertension}, volume={146}, ISSN={0007-1188 1476-5381}, url={http://dx.doi.org/10.1038/SJ.BJP.0706408}, DOI={10.1038/SJ.BJP.0706408}, abstractNote={ Inhibition of the type 5 phosphodiesterase and inhibition of Rho kinase are both effective in reducing pulmonary hypertension (PH). Here we investigate whether Rho kinase inhibition is involved in the beneficial effect of the type 5 phosphodiesterase inhibitor sildenafil on PH. Chronic hypoxia‐induced PH in rats is associated with an increase in RhoA activity in pulmonary artery that was maximal after 2 days (10.7±0.9‐fold increase, n=6, P<0.001). The activity of Rho kinase assessed by measuring the level of myosin phosphatase target subunit 1 (MYPT1) phosphorylation was also increased (5.7±0.8‐fold over control, n=8). Chronic fasudil (30 mg kg−1 day−1; 14 days) and sildenafil (25 mg kg−1 day−1; 14 days) treatments reduced PH and pulmonary cardiovascular remodelling, and inhibited the MYPT1 phosphorylation in pulmonary artery from hypoxic rats by 82.3±3% (n=4) and by 76.6±2% (n=4), respectively. The inhibitory effect of sildenafil (10 μM) on MYPT1 phosphorylation was demonstrated by the loss of actin stress fibres in vascular smooth muscle cells. However, in vitro kinase assays indicated that sildenafil had no direct inhibitory action on Rho kinase activity. Sildenafil treatment induced increased RhoA phosphorylation and association to its cytosolic inhibitory protein, guanine dissociation inhibitor (GDI) in pulmonary artery. We propose that sildenafil inhibits RhoA/Rho kinase‐dependent functions in pulmonary artery through enhanced RhoA phosphorylation and cytosolic sequestration by GDI. The inhibition of intracellular events downstream of RhoA thus participates in the beneficial effect of sildenafil on PH. }, number={7}, journal={British Journal of Pharmacology}, publisher={Wiley}, author={Guilluy, Christophe and Sauzeau, Vincent and Rolli-Derkinderen, Malvyne and Guérin, Patrice and Sagan, Christine and Pacaud, Pierre and Loirand, Gervaise}, year={2005}, month={Dec}, pages={1010–1018} }