Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ellie Tzima is active.

Publication


Featured researches published by Ellie Tzima.


Current Biology | 2012

Localized tensional forces on PECAM-1 elicit a global mechanotransduction response via the integrin-RhoA pathway.

Caitlin Collins; Christophe Guilluy; Christopher Welch; E. Timothy O’Brien; Klaus M. Hahn; Richard Superfine; Keith Burridge; Ellie Tzima

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.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2009

PECAM-1 Is Necessary for Flow-Induced Vascular Remodeling

Zhongming Chen; Ellie Tzima

Objective—Vascular remodeling is a physiological process that occurs in response to long-term changes in hemodynamic conditions, but may also contribute to the pathophysiology of intima-media thickening (IMT) and vascular disease. Shear stress detection by the endothelium is thought to be an important determinant of vascular remodeling. Previous work showed that platelet endothelial cell adhesion molecule-1 (PECAM-1) is a component of a mechanosensory complex that mediates endothelial cell (EC) responses to shear stress. Methods and Results—We tested the hypothesis that PECAM-1 contributes to vascular remodeling by analyzing the response to partial carotid artery ligation in PECAM-1 knockout mice and wild-type littermates. PECAM-1 deficiency resulted in impaired vascular remodeling and significantly reduced IMT in areas of low flow. Inward remodeling was associated with PECAM-1–dependent NF&kgr;B activation, surface adhesion molecule expression, and leukocyte infiltration as well as Akt activation and vascular cell proliferation. Conclusions—PECAM-1 plays a crucial role in the activation of the NF&kgr;B and Akt pathways and inflammatory cell accumulation during vascular remodeling and IMT. Elucidation of some of the signals that drive vascular remodeling represent pharmacologically tractable targets for the treatment of restenosis after balloon angioplasty or stent placement.


Circulation Research | 2010

Role of PECAM-1 in Arteriogenesis and Specification of Preexisting Collaterals

Zhongming Chen; Janet Rubin; Ellie Tzima

Rationale: Hemodynamic forces caused by the altered blood flow in response to an occlusion lead to the induction of collateral remodeling and arteriogenesis. Previous work showed that platelet endothelial cell adhesion molecule (PECAM)-1 is a component of a mechanosensory complex that mediates endothelial cell responses to shear stress. Objective: We hypothesized that PECAM-1 plays an important role in arteriogenesis and collateral remodeling. Methods and Results: PECAM-1 knockout (KO) and wild-type littermates underwent femoral artery ligation. Surprisingly, tissue perfusion and collateral-dependent blood flow were significantly increased in the KO mice immediately after surgery. Histology confirmed larger caliber of preexisting collaterals in the KO mice. Additionally, KO mice showed blunted recovery of perfusion from hindlimb ischemia and reduced collateral remodeling, because of deficits in shear stress–induced signaling, including activation of the nuclear factor &kgr;B pathway and inflammatory cell accumulation. Partial recovery was associated with normal responses to circumferential wall tension in the absence of PECAM-1, as evidenced by the upregulation of ephrin B2 and monocyte chemoattractant protein-1, which are 2 stretch-induced regulators of arteriogenesis, both in vitro and in vivo. Conclusions: Our findings suggest a novel role for PECAM-1 in arteriogenesis and collateral remodeling. Furthermore, we identify PECAM-1 as the first molecule that determines preexisting collateral diameter.


Nature Communications | 2014

Haemodynamic and extracellular matrix cues regulate the mechanical phenotype and stiffness of aortic endothelial cells

Caitlin Collins; Lukas D. Osborne; Christophe Guilluy; Zhongming Chen; E. Tim O’Brien; John S. Reader; Keith Burridge; Richard Superfine; Ellie Tzima

Endothelial cell (ECs) lining blood vessels express many mechanosensors, including platelet endothelial cell adhesion molecule-1 (PECAM-1), that convert mechanical force to 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 to ECs on fibronectin. This is due to PKA-dependent serine phosphorylation and inactivation of RhoA. PKA signaling regulates focal adhesion dynamics and EC compliance in response to shear stress in vitro and in vivo. Our study identifies a ECM-specific, mechanosensitive signaling pathway that regulates EC compliance and may serve as an atheroprotective mechanism maintains blood vessel integrity in vivo.


Journal of Cell Biology | 2008

Shc coordinates signals from intercellular junctions and integrins to regulate flow-induced inflammation

Yunhao Liu; Daniel Timothy Sweet; Mohamad Irani-Tehrani; Nobuyo Maeda; Ellie Tzima

Atherosclerotic plaques develop in regions of the vasculature associated with chronic inflammation due to disturbed flow patterns. Endothelial phenotype modulation by flow requires the integration of numerous mechanotransduction pathways, but how this is achieved is not well understood. We show here that, in response to flow, the adaptor protein Shc is activated and associates with cell–cell and cell–matrix adhesions. Shc activation requires the tyrosine kinases vascular endothelial growth factor receptor 2 and Src. Shc activation and its vascular endothelial cadherin (VE-cadherin) association are matrix independent. In contrast, Shc binding to integrins requires VE-cadherin but occurs only on specific matrices. Silencing Shc results in reduction in both matrix-independent and matrix-dependent signals. Furthermore, Shc regulates flow-induced inflammatory signaling by activating nuclear factor κB–dependent signals that lead to atherogenesis. In vivo, Shc is activated in atherosclerosis-prone regions of arteries, and its activation correlates with areas of atherosclerosis. Our results support a model in which Shc orchestrates signals from cell–cell and cell–matrix adhesions to elicit flow-induced inflammatory signaling.


Journal of Cell Biology | 2013

A novel pathway spatiotemporally activates Rac1 and redox signaling in response to fluid shear stress

Yunhao Liu; Caitlin Collins; William B. Kiosses; Ann M. Murray; Monika Joshi; Tyson R. Shepherd; Ernesto J. Fuentes; Ellie Tzima

The GEF Tiam1 acts as a novel molecular link to the VE-cadherin–p67phox–Par3 polarity complex, leading to localized activation of Rac1 and NADPH oxidase in response to fluid flow.


Experimental Gerontology | 2011

Hemodynamic forces in endothelial dysfunction and vascular aging

Caitlin Collins; Ellie Tzima

Aging is a key risk factor associated with the onset of cardiovascular disease. Notably, vascular aging and cardiovascular disease are both associated with endothelial dysfunction, or a marked decrease in production and bioavailability the vasodilator of nitric oxide (NO). As a result of decreased nitric oxide availability, aging vessels often exhibit endothelial cell senescence and increased oxidative stress. One of the most potent activators of NO production is fluid shear stress produced by blood flow. Interestingly, age-related decrease in NO production partially results from endothelial insensitivity to shear stress. While the endothelial cell response to fluid shear stress has been well characterized in recent years, the exact mechanisms of how the mechanical force of fluid shear stress is converted into intracellular biochemical signals are relatively unknown. Therefore, gaining a better knowledge of mechanosignaling events in endothelial cells may prove to be beneficial for developing potential therapies for cardiovascular diseases.


Vascular Pharmacology | 2010

Mammalian aminoacyl-tRNA synthetases: Cell signaling functions of the protein translation machinery

Meredith V. Brown; John S. Reader; Ellie Tzima

Aminoacyl-tRNA synthetases (aaRSs) are enzymes that join amino acids to tRNAs. Although they are housekeeping enzymes essential for protein synthesis, aaRSs are now known to participate in a wide variety of functions, including transcription, translation, splicing, inflammation, angiogenesis and apoptosis. In eukaryotes, the functional expansion of aaRSs is closely linked to evolutionary advantages conferred by recruitment into protein complexes as well as various structural adaptations. The elucidation and understanding of the diverse functions of aaRSs is a major goal of current and future research. These investigations will undoubtedly provide some of the most fundamental understanding of how and possibly why synthetases became so tightly involved in such a vast array of cell signaling pathways.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Platelet-Endothelial Cell Adhesion Molecule-1 Regulates Endothelial NO Synthase Activity and Localization Through Signal Transducers and Activators of Transcription 3–Dependent NOSTRIN Expression

Margaret E. McCormick; Reema Goel; David Fulton; Stefanie Oess; Debra K. Newman; Ellie Tzima

Background—NO produced by the endothelial NO synthase (eNOS) is an important regulator of cardiovascular physiological and pathological features. eNOS is activated by numerous stimuli, and its activity is tightly regulated. Platelet-endothelial cell adhesion molecule-1 (PECAM-1) has been implicated in regulating eNOS activity in response to shear stress. The current study was conducted to determine the role of PECAM-1 in the regulation of basal eNOS activity. Methods and Results—We demonstrate that PECAM-1–knockout ECs have increased basal eNOS activity and NO production. Mechanistically, increased eNOS activity is associated with a decrease in the inhibitory interaction of eNOS with caveolin-1, impaired subcellular localization of eNOS, and decreased eNOS traffic inducer (NOSTRIN) expression in the absence of PECAM-1. Furthermore, we demonstrate that activation of blunted signal transducers and activators of transcription 3 (STAT3) in the absence of PECAM-1 results in decreased NOSTRIN expression via direct binding of the signal transducers and activators of transcription 3 to the NOSTRIN promoter. Conclusion—Our results reveal an elegant mechanism of eNOS regulation by PECAM-1 through signal transducers and activators of transcription 3–mediated transcriptional control of NOSTRIN.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Bmper inhibits endothelial expression of inflammatory adhesion molecules and protects against atherosclerosis.

Xinchun Pi; Pamela Lockyer; Laura A. Dyer; Jonathan C. Schisler; Brooke Russell; Stephen Carey; Daniel Timothy Sweet; Zhongming Chen; Ellie Tzima; Monte S. Willis; Jonathon W. Homeister; Martin Moser; Cam Patterson

Objective—Bone morphogenetic proteins (Bmps) are important mediators of inflammation and atherosclerosis, though their mechanism of action is not fully understood. To better understand the contribution of the Bmp signaling pathway in vascular inflammation, we investigated the role of Bmper (Bmp endothelial cell precursor–derived regulator), an extracellular Bmp modulator, in an induced in vivo model of inflammation and atherosclerosis. Methods and Results—We crossed apolipoprotein E–deficient (ApoE−/−) mice with mice missing 1 allele of Bmper (Bmper+/− mice used in the place of Bmper−/− mice that die at birth) and measured the development of atherosclerosis in mice fed a high-fat diet. Bmper haploinsufficiency in ApoE−/− mice (Bmper+/−;ApoE−/− mice) led to a more severe phenotype compared with Bmper+/+;ApoE−/− mice. Bmper+/−;ApoE−/− mice also exhibited increased Bmp activity in the endothelial cells in both the greater and lesser curvatures of the aortic arch, suggesting a role for Bmper in regulating Bmp-mediated inflammation associated with laminar and oscillatory shear stress. Small interfering RNA knockdown of Bmper in human umbilical vein endothelial cells caused a dramatic increase in the inflammatory markers intracellular adhesion molecule 1 and vascular cell adhesion molecule 1 at rest and after exposure to oscillatory and laminar shear stress. Conclusion—We conclude that Bmper is a critical regulator of Bmp-mediated vascular inflammation and that the fine-tuning of Bmp and Bmper levels is essential in the maintenance of normal vascular homeostasis.

Collaboration


Dive into the Ellie Tzima's collaboration.

Top Co-Authors

Avatar

Zhongming Chen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Caitlin Collins

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Daniel Timothy Sweet

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mauricio Rojas

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Christopher S. Givens

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

John S. Reader

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

A. Phillip Owens

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Chris Givens

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Yunhao Liu

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge