Network


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

Hotspot


Dive into the research topics where Ssang-Taek Lim is active.

Publication


Featured researches published by Ssang-Taek Lim.


Molecular Cell | 2008

Nuclear FAK Promotes Cell Proliferation and Survival through FERM-Enhanced p53 Degradation

Ssang-Taek Lim; Xiao Lei Chen; Yangmi Lim; Dan A. Hanson; Thanh-Trang Vo; Kyle Howerton; Nicholas Larocque; Susan J. Fisher; David D. Schlaepfer; Dusko Ilic

FAK is known as an integrin- and growth factor-associated tyrosine kinase promoting cell motility. Here we show that, during mouse development, FAK inactivation results in p53- and p21-dependent mesodermal cell growth arrest. Reconstitution of primary FAK-/-p21-/- fibroblasts revealed that FAK, in a kinase-independent manner, facilitates p53 turnover via enhanced Mdm2-dependent p53 ubiquitination. p53 inactivation by FAK required FAK FERM F1 lobe binding to p53, FERM F2 lobe-mediated nuclear localization, and FERM F3 lobe for connections to Mdm2 and proteasomal degradation. Staurosporine or loss of cell adhesion enhanced FERM-dependent FAK nuclear accumulation. In primary human cells, FAK knockdown raised p53-p21 levels and slowed cell proliferation but did not cause apoptosis. Notably, FAK knockdown plus cisplatin triggered p53-dependent cell apoptosis, which was rescued by either full-length FAK or FAK FERM re-expression. These studies define a scaffolding role for nuclear FAK in facilitating cell survival through enhanced p53 degradation under conditions of cellular stress.


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 | 2006

Intrinsic FAK activity and Y925 phosphorylation facilitate an angiogenic switch in tumors

Satyajit K. Mitra; David Mikolon; John Molina; Datsun A. Hsia; Hanson Da; Chi A; Ssang-Taek Lim; Joie A. Bernard-Trifilo; Dusko Ilic; Dwayne G. Stupack; David A. Cheresh; David D. Schlaepfer

Elevated focal adhesion kinase (FAK) expression occurs in advanced cancers, yet a signaling role for FAK in tumor progression remains undefined. Here, we suppressed FAK activity in 4T1 breast carcinoma cells resulting in reduced FAK Y925 phosphorylation, Grb2 adaptor protein binding to FAK, and signaling to mitogen-activated protein (MAP) kinase (MAPK). Loss of a FAK-Grb2-MAPK linkage did not affect 4T1 cell proliferation or survival in culture, yet FAK inhibition reduced vascular endothelial growth factor (VEGF) expression and resulted in small avascular tumors in mice. This FAK-Grb2-MAPK linkage was essential in promoting angiogenesis as reconstitution experiments using Src-transformed FAK-null fibroblasts revealed that point mutations affecting FAK catalytic activity (R454) or Y925 phosphorylation (F925) disrupted the ability of FAK to promote MAPK- and VEGF-associated tumor growth. Notably, in both FAK-inhibited 4T1 and Src-transformed FAK-null cells, constitutively activated (CA) mitogen-activated protein kinase kinase 1 (MEK1) restored VEGF production and CA-MEK1 or added VEGF rescued tumor growth and angiogenesis. These studies provide the first biological support for Y925 FAK phosphorylation and define a novel role for FAK activity in promoting a MAPK-associated angiogenic switch during tumor progression.


Journal of Cell Biology | 2008

Compensatory role for Pyk2 during angiogenesis in adult mice lacking endothelial cell FAK

Sara M. Weis; Ssang-Taek Lim; Kimberly Lutu-Fuga; Leo Barnes; Xiao Lei Chen; Joachim R. Göthert; Tang-Long Shen; Jun-Lin Guan; David D. Schlaepfer; David A. Cheresh

Focal adhesion kinase (FAK) plays a critical role during vascular development because knockout of FAK in endothelial cells (ECs) is embryonic lethal. Surprisingly, tamoxifen-inducible conditional knockout of FAK in adult blood vessels (inducible EC–specific FAK knockout [i-EC-FAK-KO]) produces no vascular phenotype, and these animals are capable of developing a robust growth factor–induced angiogenic response. Although angiogenesis in wild-type mice is suppressed by pharmacological inhibition of FAK, i-EC-FAK-KO mice are refractory to this treatment, which suggests that adult i-EC-FAK-KO mice develop a compensatory mechanism to bypass the requirement for FAK. Indeed, expression of the FAK-related proline-rich tyrosine kinase 2 (Pyk2) is elevated and phosphorylated in i-EC-FAK-KO blood vessels. In cultured ECs, FAK knockdown leads to increased Pyk2 expression and, surprisingly, FAK kinase inhibition leads to increased Pyk2 phosphorylation. Pyk2 can functionally compensate for the loss of FAK because knockdown or pharmacological inhibition of Pyk2 disrupts angiogenesis in i-EC-FAK-KO mice. These studies reveal the adaptive capacity of ECs to switch to Pyk2-dependent signaling after deletion or kinase inhibition of FAK.


Journal of Cell Science | 2009

A FAK-p120RasGAP-p190RhoGAP complex regulates polarity in migrating cells

Alok Tomar; Ssang-Taek Lim; Yangmi Lim; David D. Schlaepfer

Directional motility is a complex process requiring the spatiotemporal integration of signals that regulate cytoskeletal changes, and the establishment of an anteroposterior or polarized cell axis. Focal adhesion kinase (FAK) promotes cell migration, but a molecular role for FAK in promoting cell polarity remains undefined. Here, using wound healing and Golgi-reorientation analyses, we show that fibroblast, endothelial and carcinoma polarity during cell migration requires FAK and is associated with a complex between FAK, p120RasGAP and p190RhoGAP (p190A), leading to p190A tyrosine phosphorylation. Fibronectin-integrin-mediated FAK activation and phosphorylation promote SH2-mediated binding of p120RasGAP to FAK and FAK-mediated p190A tyrosine phosphorylation. The association of p120RasGAP with FAK facilitates the formation of a FAK-p120RasGAP-p190A complex targeted to leading-edge focal adhesions by FAK. Knockdown of p120RasGAP, mutation of FAK Y397 or inhibition of FAK activity prevent the association of FAK with p190A and subsequent tyrosine phosphorylation of p190A, and result in the loss of cell polarity. Because reconstitution of FAK-null fibroblasts with FAK or a Pyk2-FAK chimera restore the normal decrease in RhoA GTP binding upon cell spreading on fibronectin, our studies support a model whereby FAK activity facilitates the recruitment and stabilization of a p120RasGAP-p190A complex at leading-edge focal adhesions connected to the transient inhibition of RhoA activity and the regulation of cell polarity.


Cell Cycle | 2008

FERM control of FAK function: Implications for cancer therapy

Ssang-Taek Lim; David Mikolon; Dwayne G. Stupack; David D. Schlaepfer

Integrins are transmembrane receptors that bind to extracellular matrix proteins and convey anchorage-dependent signals regulating normal cell proliferation. Integrin signals within the tumor micro-environment also impact cancer cell survival and invasion during tumor progression. These integrin-associated signaling events are transduced in part through the activation of non-receptor protein-tyrosine kinases. Focal adhesion kinase (FAK) is activated by β-subunit integrins in both normal and transformed cells. As genetic inactivation of β1 integrin or FAK yield early embryonic lethal phenotypes associated with decreased cell proliferation, and dominant-negative inhibition of FAK can cause increased cell apoptosis, there is a concern that FAK inhibition may have cytotoxic effects on cell growth or survival.However, FAK-specific small molecule inhibitors do not directly impact cell growth in culture, but yet show potent anti-tumor growth effects in vivo. Additionally, recent studies have shed new insight into the FAK kinase-independent regulation of cell proliferation and survival mediated by the FAK N-terminal FERM (band 4.1, ezrin, radixin, moesin homology) domain.Herein, we review the role of the FAK FERM domain in both the intrinsic regulation of FAKkinase activity and how FERM-mediated nuclear localization of FAK promotes enhanced cell survival through the inhibition of tumor suppressor p53 activation during development and under conditions of cellular stress. As we find that FAK FERM-mediated regulation of p53 occurs in human carcinoma cells, elevated FAK expression in tumors may promote both kinase-dependent and –independent survival mechanisms. We discuss how the pharmacological inhibition of FAK kinase activity may impact tumor progression through combined effects of blocking both tumor- and stromal-associated signaling regulating neovascularization.


Oncogene | 2006

Intrinsic focal adhesion kinase activity controls orthotopic breast carcinoma metastasis via the regulation of urokinase plasminogen activator expression in a syngeneic tumor model.

Satyajit K. Mitra; Ssang-Taek Lim; Chi A; David D. Schlaepfer

Expression of focal adhesion kinase (FAK) is elevated in malignant breast cancer, yet the role of intrinsic FAK activity in promoting tumor progression remains undefined. Here, we have inhibited FAK activity or expression in murine 4T1 breast carcinoma cells via dominant-negative focal adhesion kinase-related non-kinase (FRNK) or anti-FAK short hairpin RNA (shRNA) expression, respectively. Neither FRNK nor FAK shRNA (∼80% reduced FAK levels) affected 4T1 proliferation in culture, whereas reduced FAK activity or expression blocked 4T1 cell invasion through Matrigel and resulted in 2–3-fold lower urokinase plasminogen activator (uPA) expression. Control 4T1 cells implanted into mammary fat pads of BALB/c mice exhibited spontaneous metastasis to the lungs, to the peritoneal cavity, and resulted in 90% lethality within 21 days. Whereas FAK shRNA-expressing 4T1 cells formed tumors in mice with low levels of apoptosis, when mammary-injected, these cells did not exhibit lung metastasis after 21 days and caused only 40% lethality up to 60 days. Transient re-expression of wild-type but not kinase-dead FAK in 4T1 FAK shRNA cells promoted uPA production and mammary to lung metastasis within 7 days. In fact, stable human uPA overexpression in 4T1 FAK shRNA cells promoted Matrigel invasion and lung metastasis equal to 4T1 controls. Conversely, treatment with plasminogen activator inhibitor-1 or neutralizing antibody to uPA blocked Matrigel invasion of 4T1 control cells. These studies provide the first direct proof that FAK catalytic activity can facilitate metastatic breast cancer progression by regulating uPA expression.


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.

Collaboration


Dive into the Ssang-Taek Lim's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiao Lei Chen

University of California

View shared research outputs
Top Co-Authors

Avatar

Alok Tomar

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Sean Uryu

University of California

View shared research outputs
Top Co-Authors

Avatar

Yangmi Lim

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ju-Ock Nam

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Colin Walsh

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge