Network


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

Hotspot


Dive into the research topics where Sean Uryu is active.

Publication


Featured researches published by Sean Uryu.


Developmental Cell | 2012

VEGF-Induced Vascular Permeability Is Mediated by FAK

Xiao Lei Chen; Ju-Ock Nam; Christine Jean; Christine Lawson; Colin Walsh; Erik Goka; Ssang-Taek Lim; Alok Tomar; Isabelle Tancioni; Sean Uryu; Jun-Lin Guan; Lisette M. Acevedo; Sara M. Weis; David A. Cheresh; David D. Schlaepfer

Endothelial cells (ECs) form cell-cell adhesive junctional structures maintaining vascular integrity. This barrier is dynamically regulated by vascular endothelial growth factor (VEGF) receptor signaling. We created an inducible knockin mouse model to study the contribution of the integrin-associated focal adhesion tyrosine kinase (FAK) signaling on vascular function. Here we show that genetic or pharmacological FAK inhibition in ECs prevents VEGF-stimulated permeability downstream of VEGF receptor or Src tyrosine kinase activation in vivo. VEGF promotes tension-independent FAK activation, rapid FAK localization to cell-cell junctions, binding of the FAK FERM domain to the vascular endothelial cadherin (VE-cadherin) cytoplasmic tail, and direct FAK phosphorylation of β-catenin at tyrosine-142 (Y142) facilitating VE-cadherin-β-catenin dissociation and EC junctional breakdown. Kinase inhibited FAK is in a closed conformation that prevents VE-cadherin association and limits VEGF-stimulated β-catenin Y142 phosphorylation. Our studies establish a role for FAK as an essential signaling switch within ECs regulating adherens junction dynamics.


Journal of Cell Biology | 2008

PyK2 and FAK connections to p190Rho guanine nucleotide exchange factor regulate RhoA activity, focal adhesion formation, and cell motility

Yangmi Lim; Ssang-Taek Lim; Alok Tomar; Margaret L. Gardel; Joie A. Bernard-Trifilo; Xiao Lei Chen; Sean Uryu; Rafaela Cañete-Soler; Jinbin Zhai; Hong Lin; William W. Schlaepfer; Perihan Nalbant; Gary M. Bokoch; Dusko Ilic; Clare M. Waterman-Storer; David D. Schlaepfer

Integrin binding to matrix proteins such as fibronectin (FN) leads to formation of focal adhesion (FA) cellular contact sites that regulate migration. RhoA GTPases facilitate FA formation, yet FA-associated RhoA-specific guanine nucleotide exchange factors (GEFs) remain unknown. Here, we show that proline-rich kinase-2 (Pyk2) levels increase upon loss of focal adhesion kinase (FAK) in mouse embryonic fibroblasts (MEFs). Additionally, we demonstrate that Pyk2 facilitates deregulated RhoA activation, elevated FA formation, and enhanced cell proliferation by promoting p190RhoGEF expression. In normal MEFs, p190RhoGEF knockdown inhibits FN-associated RhoA activation, FA formation, and cell migration. Knockdown of p190RhoGEF-related GEFH1 does not affect FA formation in FAK−/− or normal MEFs. p190RhoGEF overexpression enhances RhoA activation and FA formation in MEFs dependent on FAK binding and associated with p190RhoGEF FA recruitment and tyrosine phosphorylation. These studies elucidate a compensatory function for Pyk2 upon FAK loss and identify the FAK–p190RhoGEF complex as an important integrin-proximal regulator of FA formation during FN-stimulated cell motility.


Journal of Cell Biology | 2012

FAK promotes recruitment of talin to nascent adhesions to control cell motility

Christine Lawson; Ssang-Taek Lim; Sean Uryu; Xiao Lei Chen; David A. Calderwood; David D. Schlaepfer

An alternative linkage is shown whereby FAK brings talin to nascent adhesions independent of talin binding to β1 integrins.


Oncogene | 2008

Distinct FAK-Src activation events promote α5β1 and α4β1 integrin-stimulated neuroblastoma cell motility

Lihua Wu; Joie A. Bernard-Trifilo; Yangmi Lim; Ssang-Taek Lim; Satyajit K. Mitra; Sean Uryu; Min Chen; Catherine J. Pallen; Nai-Kong V. Cheung; David Mikolon; Ainhoa Mielgo; Dwayne G. Stupack; David D. Schlaepfer

Signals from fibronectin-binding integrins promote neural crest cell motility during development in part through protein-tyrosine kinase (PTK) activation. Neuroblastoma (NB) is a neural crest malignancy with high metastatic potential. We find that α4 and α5 integrins are present in late-stage NB tumors and cell lines derived thereof. To determine the signaling connections promoting either α4β1- or α5β1-initiated NB cell motility, pharmacological, dominant negative and short-hairpin RNA (shRNA) inhibitory approaches were undertaken. shRNA knockdown revealed that α5β1-stimulated NB motility is dependent upon focal adhesion kinase (FAK) PTK, Src PTK and p130Cas adapter protein expression. Cell reconstitution showed that FAK catalytic activity is required for α5β1-stimulated Src activation in part through direct FAK phosphorylation of Src at Tyr-418. Alternatively, α4β1-stimulated NB cell motility is dependent upon Src and p130Cas but FAK is not essential. Catalytically inactive receptor protein-tyrosine phosphatase-α overexpression inhibited α4β1-stimulated NB motility and Src activation consistent with α4-regulated Src activity occurring through Src Tyr-529 dephosphorylation. In α4 shRNA-expressing NB cells, α4β1-stimulated Src activation and NB cell motility were rescued by wild type but not cytoplasmic domain-truncated α4 re-expression. These studies, supported by results using reconstituted fibroblasts, reveal that α4β1-mediated Src activation is mechanistically distinct from FAK-mediated Src activation during α5β1-mediated NB migration and support the evaluation of inhibitors to α4, Src and FAK in the control of NB tumor progression.


Cancer Biology & Therapy | 2010

PND-1186 FAK inhibitor selectively promotes tumor cell apoptosis in three-dimensional environments.

Isabelle Tanjoni; Colin Walsh; Sean Uryu; Alok Tomar; Ju-Ock Nam; Ainhoa Mielgo; Ssang-Taek Lim; Congxin Liang; Marcel Koenig; Neela Patel; Cheni Kwok; Gerald McMahon; Dwayne G. Stupack; David D. Schlaepfer

Tumor cells can grow in an anchorage-independent manner. This is mediated in part through survival signals that bypass normal growth restraints controlled by integrin cell surface receptors. Focal adhesion kinase (FAK) is a cytoplasmic protein-tyrosine kinase that associates with integrins and modulates various cellular processes including growth, survival, and migration. As increased FAK expression and tyrosine phosphorylation are associated with tumor progression, inhibitors of FAK are being tested for anti-tumor effects. Here, we analyze PND-1186, a substituted pyridine reversible inhibitor of FAK activity with a 50% inhibitory concentration (IC50) of 1.5 nM in vitro. PND-1186 has an IC50 of ~100 nM in breast carcinoma cells as determined by anti-phospho-specific immunoblotting to FAK Tyr-397. PND-1186 did not alter c‑Src or p130Cas tyrosine phosphorylation in adherent cells, yet functioned to restrain cell movement. Notably, 1.0 µM PND-1186 (>5-fold above IC50) had limited effects on cell proliferation. However, under non-adherent conditions as spheroids and as colonies in soft agar, 0.1 µM PND-1186 blocked FAK and p130Cas tyrosine phosphorylation, promoted caspase-3 activation, and triggered cell apoptosis. PND-1186 inhibited 4T1 breast carcinoma subcutaneous tumor growth correlated with elevated tumor cell apoptosis and caspase 3 activation. Addition of PND-1186 to the drinking water of mice was well tolerated and inhibited ascites- and peritoneal membrane-associated ovarian carcinoma tumor growth associated with the inhibition of FAK Tyr-397 phosphorylation. Our results with low-level PND-1186 treatment support the conclusion that FAK activity selectively promotes tumor cell survival in three-dimensional environments.


Cancer Biology & Therapy | 2010

Oral delivery of PND-1186 FAK inhibitor decreases tumor growth and spontaneous breast to lung metastasis in pre-clinical models

Colin Walsh; Isabelle Tanjoni; Sean Uryu; Alok Tomar; Ju-Ock Nam; Hong Luo; Angelica Phillips; Neela Patel; Cheni Kwok; Gerald McMahon; Dwayne G. Stupack; David D. Schlaepfer

Tumor metastasis is a leading cause of cancer-related death. Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase recruited to integrin-mediated matrix attachment sites where FAK activity is implicated in the control of cell survival, migration, and invasion. Although genetic studies support the importance of FAK activity in promoting tumor progression, it remains unclear whether pharmacological FAK inhibition prevents tumor metastasis. Here, we show that the FAK inhibitor PND-1186 blocks FAK Tyr-397 phosphorylation in vivo and exhibits anti-tumor efficacy in orthotopic breast carcinoma mouse tumor models. PND-1186 (100 mg/kg intraperitoneal, i.p.) showed promising pharmacokinetics (PK) and inhibited tumor FAK Tyr-397 phosphorylation for 12 hours. Oral administration of 150 mg/kg PND-1186 gave a more sustained PK profile verses i.p., and when given twice daily, PND-1186 significantly inhibited sygeneic murine 4T1 orthotopic breast carcinoma tumor growth and spontaneous metastasis to lungs. Moreover, low-level 0.5 mg/ml PND-1186 ad libitum administration in drinking water prevented oncogenic KRAS- and BRAF-stimulated MDA-MB-231 breast carcinoma tumor growth and metastasis with inhibition of tumoral FAK and p130Cas phosphorylation. Although PND-1186 was not cytotoxic to cells in adherent culture, tumors from animals receiving PND-1186 exhibited increased TUNEL staining, decreased leukocyte infiltrate and reduced tumor-associated splenomegaly. In vitro, PND-1186 reduced tumor necrosis factor-a triggered interleukin-6 cytokine expression, indicating that FAK inhibition may impact tumor progression via effects on both tumor and stromal cells. As oral administration of PND-1186 also decreased experimental tumor metastasis, PND-1186 may therefore be useful clinically to curb breast tumor progression.


Journal of Cell Biology | 2014

Inhibition of endothelial FAK activity prevents tumor metastasis by enhancing barrier function

Christine Jean; Xiao Lei Chen; Ju-Ock Nam; Isabelle Tancioni; Sean Uryu; Christine Lawson; Kristy K. Ward; Colin Walsh; Nichol L. G. Miller; Majid Ghassemian; Patric Turowski; Elisabetta Dejana; Sara M. Weis; David A. Cheresh; David D. Schlaepfer

Endothelial cell focal adhesion kinase is a key intermediate between c-Src and the regulation of endothelial cell barrier function in the control of tumor metastasis.


Journal of Cell Biology | 2012

Nuclear-localized focal adhesion kinase regulates inflammatory VCAM-1 expression

Ssang-Taek Lim; Nichol L. G. Miller; Xiao Lei Chen; Isabelle Tancioni; Colin Walsh; Christine Lawson; Sean Uryu; Sara M. Weis; David A. Cheresh; David D. Schlaepfer

Kinase-inhibited FAK limits VCAM-1 production via nuclear localization and promotion of GATA4 turnover.


Journal of Biological Chemistry | 2007

Tumor necrosis factor-alpha stimulates focal adhesion kinase activity required for mitogen-activated kinase-associated interleukin 6 expression.

David D. Schlaepfer; Shihe Hou; Ssang-Taek Lim; Alok Tomar; Honggang Yu; Yangmi Lim; Dan A. Hanson; Sean Uryu; John Molina; Satyajit K. Mitra

Focal adhesion kinase (FAK) is a cytoplasmic protein-tyrosine kinase that promotes cell migration, survival, and gene expression. Here we show that FAK signaling is important for tumor necrosis factor-α (TNFα)-induced interleukin 6 (IL-6) mRNA and protein expression in breast (4T1), lung (A549), prostate (PC-3), and neural (NB-8) tumor cells by FAK short hairpin RNA knockdown and by comparisons of FAK-null (FAK–/–) and FAK+/+ mouse embryo fibroblasts. FAK promoted TNFα-stimulated MAPK activation needed for maximal IL-6 production. FAK was not required for TNFα-mediated nuclear factor-κB or c-Jun N-terminal kinase activation. TNFα-stimulated FAK catalytic activation and IL-6 production were inhibited by FAK N-terminal but not FAK C-terminal domain overexpression. Analysis of FAK–/– fibroblasts stably reconstituted with wild type or various FAK point mutants showed that FAK catalytic activity, Tyr-397 phosphorylation, and the Pro-712/713 proline-rich region of FAK were required for TNFα-stimulated MAPK activation and IL-6 production. Constitutively activated MAPK kinase-1 (MEK1) expression in FAK–/– and A549 FAK short hairpin RNA-expressing cells rescued TNFα-stimulated IL-6 production. Inhibition of Src protein-tyrosine kinase activity or mutation of Src phosphorylation sites on FAK (Tyr-861 or Tyr-925) did not affect TNFα-stimulated IL-6 expression. Moreover, analyses of Src–/–, Yes–/–, and Fyn–/– fibroblasts showed that Src expression was inhibitory to TNFα-stimulated IL-6 production. These studies provide evidence for a novel Src-independent FAK to MAPK signaling pathway regulating IL-6 expression with potential importance to inflammation and tumor progression.


Journal of Biological Chemistry | 2007

Tumor necrosis factor-α stimulates FAK activity required for mitogen-activated kinase-associated interleukin 6 expression

David D. Schlaepfer; Shihe Hou; Ssang-Taek Lim; Alok Tomar; Honggang Yu; Yangmi Lim; Dan A. Hanson; Sean Uryu; John Molina; Satyajit K. Mitra

Focal adhesion kinase (FAK) is a cytoplasmic protein-tyrosine kinase that promotes cell migration, survival, and gene expression. Here we show that FAK signaling is important for tumor necrosis factor-α (TNFα)-induced interleukin 6 (IL-6) mRNA and protein expression in breast (4T1), lung (A549), prostate (PC-3), and neural (NB-8) tumor cells by FAK short hairpin RNA knockdown and by comparisons of FAK-null (FAK–/–) and FAK+/+ mouse embryo fibroblasts. FAK promoted TNFα-stimulated MAPK activation needed for maximal IL-6 production. FAK was not required for TNFα-mediated nuclear factor-κB or c-Jun N-terminal kinase activation. TNFα-stimulated FAK catalytic activation and IL-6 production were inhibited by FAK N-terminal but not FAK C-terminal domain overexpression. Analysis of FAK–/– fibroblasts stably reconstituted with wild type or various FAK point mutants showed that FAK catalytic activity, Tyr-397 phosphorylation, and the Pro-712/713 proline-rich region of FAK were required for TNFα-stimulated MAPK activation and IL-6 production. Constitutively activated MAPK kinase-1 (MEK1) expression in FAK–/– and A549 FAK short hairpin RNA-expressing cells rescued TNFα-stimulated IL-6 production. Inhibition of Src protein-tyrosine kinase activity or mutation of Src phosphorylation sites on FAK (Tyr-861 or Tyr-925) did not affect TNFα-stimulated IL-6 expression. Moreover, analyses of Src–/–, Yes–/–, and Fyn–/– fibroblasts showed that Src expression was inhibitory to TNFα-stimulated IL-6 production. These studies provide evidence for a novel Src-independent FAK to MAPK signaling pathway regulating IL-6 expression with potential importance to inflammation and tumor progression.

Collaboration


Dive into the Sean Uryu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiao Lei Chen

University of California

View shared research outputs
Top Co-Authors

Avatar

Christine Jean

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ssang-Taek Lim

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge