Network


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

Hotspot


Dive into the research topics where Kristen L. Leslie is active.

Publication


Featured researches published by Kristen L. Leslie.


Mutation Research-genetic Toxicology and Environmental Mutagenesis | 2011

Characterization and detection of cellular and proteomic alterations in stable stathmin-overexpressing, taxol-resistant BT549 breast cancer cells using offgel IEF/PAGE difference gel electrophoresis

Manimalha Balasubramani; Chitose Nakao; Guy Uechi; John J. Cardamone; Kathy Kamath; Kristen L. Leslie; Raghavan Balachandran; Leslie Wilson; Billy W. Day; Mary Ann Jordan

Stathmin/oncoprotein 18, a protein that regulates microtubule dynamics, is highly expressed in a number of tumors including leukemia, lymphoma, neuroblastoma, breast, ovarian, and prostate cancers. High stathmin levels have been associated with the development of resistance to the widely used anti-cancer drug taxol ((®)Taxol, paclitaxel). The mechanisms of stathmin-mediated taxol resistance are not well-understood at the molecular level. To better understand the role of stathmin in taxol resistance, we stably overexpressed stathmin twofold in BT549 human breast cancer cells and characterized several cell processes involved in the mechanism of action of taxol. After stable overexpression of stathmin, neither the cell doubling time nor the mitotic index was altered and the microtubule polymer mass was reduced only modestly (by 18%). Unexpectedly, microtubule dynamicity was reduced by 29% after stathmin overexpression, resulting primarily from reduction in the catastrophe frequency. Sensitivity to taxol was reduced significantly (by 44%) in a clonogenic assay, and stathmin appeared to protect the cells from the spindle-damaging effects of taxol. The results suggest that in the stably stathmin-overexpressing clones, compensatory gene expression occurred that resulted in normal rates of cell proliferation and prevented the increase in catastrophe frequency expected in response to stathmin. Stathmin overexpression protected the cells from taxol-induced abnormal mitoses, and thus induced taxol resistance. Using offgel IEF/PAGE difference gel electrophoresis, we identified a number of proteins whose expression is reduced in the taxol-resistant stathmin-overexpressing cell lines, including proteins involved in the cytoskeleton and cell structure, the stress response, protein folding, glycolysis, and catalysis.


Journal of Molecular and Cellular Cardiology | 2010

EBP50 inhibits the anti-mitogenic action of the parathyroid hormone type 1 receptor in vascular smooth muscle cells

Gyun Jee Song; Stacey Barrick; Kristen L. Leslie; Brian M. Sicari; Nathalie Fiaschi-Taesch; Alessandro Bisello

Parathyroid hormone-related protein (PTHrP) and the parathyroid hormone type 1 receptor (PTH1R) are important regulators of vascular remodeling. PTHrP expression is associated to increased proliferation of vascular smooth muscle cells (VSMC). In contrast, signaling via the PTH1R inhibits cell growth. The mechanisms regulating the dual effect of PTHrP and PTH1R on VSMC proliferation are only partially understood. In this study we examined the role of the adaptor protein ezrin-radixin-moesin-binding phosphoprotein (EBP50) on PTH1R expression, trafficking, signaling and control of A10 cell proliferation. In normal rat vascular tissues, EBP50 was restricted to the endothelium with little expression in VSMC. EBP50 expression significantly increased in VSMC following angioplasty in parallel with PTHrP. Interestingly, PTHrP was able to induce EBP50 expression. In the clonal rat aortic smooth muscle cell line A10, EBP50 increased the recruitment of PTH1R to the cell membrane and delayed its internalization in response to PTHrP(1-36). This effect required an intact C-terminal motif in the PTH1R. In naïve A10 cells, PTHrP(1-36) stimulated cAMP production but not intracellular calcium release. In contrast, PTHrP(1-36) induced both cAMP and calcium signaling in A10 cells over-expressing EBP50. Finally, EBP50 attenuated the induction of p27(kip1) and the anti-proliferative effect of PTHrP(1-36). In summary, this study demonstrates the dynamic expression of EBP50 in vessels following injury and the effects of EBP50 on PTH1R function in VSMC. These findings highlight one of the mechanisms leading to increased VSMC proliferation and have important implication in the understanding of the molecular events leading to restenosis.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

The scaffolding protein EBP50 promotes vascular smooth muscle cell proliferation and neointima formation by regulating Skp2 and p21(cip1).

Gyun Jee Song; Stacey Barrick; Kristen L. Leslie; Philip M. Bauer; Verónica Alonso; Peter A. Friedman; Nathalie Fiaschi-Taesch; Alessandro Bisello

Objective— The Ezrin-radixin–moesin–binding phosphoprotein 50 (EBP50) is a scaffolding protein known to regulate ion homeostasis in the kidney and intestine. Previous work showed that EBP50 expression increases after balloon injury in rat carotids. This study was designed to determine the role of EBP50 on vascular smooth muscle cells (VSMC) proliferation and the development of neointimal hyperplasia. Methods and Results— Wire injury was performed in wild type (WT) and EBP50 knockout (KO) mice. Two weeks after injury, neointima formation was 80% lower in KO than in WT mice. Proliferation of KO VSMC was significantly lower than WT cells and overexpression of EBP50 increased VSMC proliferation. Akt activity and expression of S-phase kinase protein2 decreased in KO cells resulting in the stabilization of the cyclin-dependent kinase inhibitor, p21cip1. Consequently, KO cells were arrested in G0/G1 phase. Consistent with these observations, p21cip1 was detected in injured femoral arteries of KO but not WT mice. No differences in apoptosis between WT and KO were observed. Conclusion— EBP50 is critical for neointima formation and induces VSMC proliferation by decreasing S-phase kinase protein2 stability, thereby accelerating the degradation of the cell cycle inhibitor p21cip1.


Journal of Molecular and Cellular Cardiology | 2012

EBP50 Promotes Focal Adhesion Turnover and Vascular Smooth Muscle Cells Migration

Gyun Jee Song; Kristen L. Leslie; Stacey Barrick; Sylvain Bougoin; Juan M. Taboas; Alessandro Bisello

The ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) is a PDZ-containing scaffolding protein that regulates a variety of physiological functions. In the vasculature, EBP50 promotes neointima formation following arterial injury. In this study the role of EBP50 on vascular smooth muscle cell (VSMC) migration was characterized. The spreading and motility of primary VSMC isolated from EBP50 knockout (KO) mice were significantly reduced compared to wild-type (WT) cells. EBP50-null VSMC had fewer and larger focal adhesions than wild-type cells. Assembly and disassembly of focal adhesion-assessed by live-cell total internal reflection fluorescence imaging-in response to epidermal growth factor (EGF) were significantly reduced in KO cells. Immunoprecipitation experiments showed that EBP50 interacts with EGF receptor via the PDZ2 domain and with focal adhesion kinase (FAK) via the C-terminal ERM domain. EBP50 promoted the formation of a complex containing both EGF receptor and FAK. Phosphorylation of Tyr-925 of FAK in response to EGF was significantly reduced in KO cell compared to WT cells. The residence time of FAK in focal adhesions-determined by fluorescence recovery after photobleaching-was increased in WT cells. Collectively, these studies indicate that EBP50, by scaffolding EGF receptor and FAK, facilitates activation of FAK, focal adhesion turnover, and migration of VSMC.


Journal of Biological Chemistry | 2013

Ezrin-Radixin-Moesin-binding Phosphoprotein 50 (EBP50) and Nuclear Factor-κB (NF-κB) A FEED-FORWARD LOOP FOR SYSTEMIC AND VASCULAR INFLAMMATION

Kristen L. Leslie; Gyun Jee Song; Stacey Barrick; Vanessa L. Wehbi; Jean-Pierre Vilardaga; Philip M. Bauer; Alessandro Bisello

Background: EBP50 is a scaffolding protein that is critical during vascular remodeling. Results: EBP50 expression is induced by inflammatory stimuli and potentiates NF-κB activation and inflammation. Conclusion: EBP50 and NF-κB participate in a feed-forward loop leading to increased macrophage activation and enhanced response of vascular cells to inflammation. Significance: This work reveals a novel mechanism regulating systemic and vascular inflammation. The interaction between vascular cells and macrophages is critical during vascular remodeling. Here we report that the scaffolding protein, ezrin-binding phosphoprotein 50 (EBP50), is a central regulator of macrophage and vascular smooth muscle cells (VSMC) function. EBP50 is up-regulated in intimal VSMC following endoluminal injury and promotes neointima formation. However, the mechanisms underlying these effects are not fully understood. Because of the fundamental role that inflammation plays in vascular diseases, we hypothesized that EBP50 mediates macrophage activation and the response of vessels to inflammation. Indeed, EBP50 expression increased in primary macrophages and VSMC, and in the aorta of mice, upon treatment with LPS or TNFα. This increase was nuclear factor-κB (NF-κB)-dependent. Conversely, activation of NF-κB was impaired in EBP50-null VSMC and macrophages. We found that inflammatory stimuli promote the formation of an EBP50-PKCζ complex at the cell membrane that induces NF-κB signaling. Macrophage activation and vascular inflammation after acute LPS treatment were reduced in EBP50-null cells and mice as compared with WT. Furthermore, macrophage recruitment to vascular lesions was significantly reduced in EBP50 knock-out mice. Thus, EBP50 and NF-κB participate in a feed-forward loop leading to increased macrophage activation and enhanced response of vascular cells to inflammation.


Journal of Biological Chemistry | 2015

Phosphorylation of Ezrin-Radixin-Moesin-binding Phosphoprotein 50 (EBP50) by Akt Promotes Stability and Mitogenic Function of S-phase Kinase-associated Protein-2 (Skp2)

Gyun Jee Song; Kristen L. Leslie; Stacey Barrick; Tatyana Mamonova; Jeremy M. Fitzpatrick; Kenneth W. Drombosky; Noah Peyser; Bin Wang; Maria Pellegrini; Philip M. Bauer; Peter A. Friedman; Dale F. Mierke; Alessandro Bisello

Background: The PDZ scaffolding protein EBP50 promotes Akt-dependent cell proliferation through the S-phase kinase associated protein-2 Skp2. Results: Akt phosphorylates threonine 156 of EBP50, allosterically promoting the binding of Skp2 to the first PDZ domain of EBP50. Conclusion: The interaction with EBP50 regulates the localization and stability of Skp2 and promotes cell proliferation. Significance: These studies define a regulatory mechanism contributing to Akt-dependent cell proliferation. The regulation of the cell cycle by the ubiquitin-proteasome system is dependent on the activity of E3 ligases. Skp2 (S-phase kinase associated protein-2) is the substrate recognition subunit of the E3 ligase that ubiquitylates the cell cycle inhibitors p21cip1 and p27kip1 thus promoting cell cycle progression. Increased expression of Skp2 is frequently observed in diseases characterized by excessive cell proliferation, such as cancer and neointima hyperplasia. The stability and cellular localization of Skp2 are regulated by Akt, but the molecular mechanisms underlying these effects remain only partly understood. The scaffolding protein Ezrin-Binding Phosphoprotein of 50 kDa (EBP50) contains two PDZ domains and plays a critical role in the development of neointimal hyperplasia. Here we report that EBP50 directly binds Skp2 via its first PDZ domain. Moreover, EBP50 is phosphorylated by Akt on Thr-156 within the second PDZ domain, an event that allosterically promotes binding to Skp2. The interaction with EBP50 causes cytoplasmic localization of Skp2, increases Skp2 stability and promotes proliferation of primary vascular smooth muscle cells. Collectively, these studies define a novel regulatory mechanism contributing to aberrant cell growth and highlight the importance of scaffolding function of EBP50 in Akt-dependent cell proliferation.


Circulation | 2013

Abstract 13042: Ezrin Binding Phosphoprotein 50 Promotes Inflammation and Atherosclerosis

Kristen L. Leslie; Gyun Jee Song; Stacey Barrick; Vanessa L. Wehbi; Jean Pierre Vilardaga; Philip M. Bauer; Alessandro Bisello


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Abstract 32: EBP50 and NF-κB: A Feed-Forward Regulation of Vascular Inflammation

Kristen L. Leslie; GyunJee Song; Stacey Barrick; Alessandro Bisello


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Abstract 362: Akt-Dependent Interaction with EBP50 Regulates Skp2 Localization and Stability

Gyun Jee Song; Stacey Barrick; Kristen L. Leslie; Alessandro Bisello


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Abstract 487: EBP50 Promotes Epidermal Growth Factor-Dependent Activation of Focal Adhesion Kinase and Vascular Smooth Muscle Cells Migration

Gyun Jee Song; Kristen L. Leslie; Stacey Barrick; Sylvain Bougoin; Alessandro Bisello

Collaboration


Dive into the Kristen L. Leslie's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stacey Barrick

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Gyun Jee Song

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Billy W. Day

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Bin Wang

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge