Network


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

Hotspot


Dive into the research topics where Shaoying Lu is active.

Publication


Featured researches published by Shaoying Lu.


Journal of Cellular Physiology | 2009

Substrate rigidity regulates Ca2+ oscillation via RhoA pathway in stem cells.

Tae-Jin Kim; Jihye Seong; Mingxing Ouyang; Jie Sun; Shaoying Lu; Jun Pyu Hong; Ning Wang; Yingxiao Wang

Substrate rigidity plays crucial roles in regulating cellular functions, such as cell spreading, traction forces, and stem cell differentiation. However, it is not clear how substrate rigidity influences early cell signaling events such as calcium in living cells. Using highly sensitive Ca2+ biosensors based on fluorescence resonance energy transfer (FRET), we investigated the molecular mechanism by which substrate rigidity affects calcium signaling in human mesenchymal stem cells (HMSCs). Spontaneous Ca2+ oscillations were observed inside the cytoplasm and the endoplasmic reticulum (ER) using the FRET biosensors targeted at subcellular locations in cells plated on rigid dishes. Lowering the substrate stiffness to 1 kPa significantly inhibited both the magnitudes and frequencies of the cytoplasmic Ca2+ oscillation in comparison to stiffer or rigid substrate. This Ca2+ oscillation was shown to be dependent on ROCK, a downstream effector molecule of RhoA, but independent of actin filaments, microtubules, myosin light chain kinase, or myosin activity. Lysophosphatidic acid, which activates RhoA, also inhibited the frequency of the Ca2+ oscillation. Consistently, either a constitutive active mutant of RhoA (RhoA‐V14) or a dominant negative mutant of RhoA (RhoA‐N19) inhibited the Ca2+ oscillation. Further experiments revealed that HMSCs cultured on gels with low elastic moduli displayed low RhoA activities. Therefore, our results demonstrate that RhoA and its downstream molecule ROCK may mediate the substrate rigidity‐regulated Ca2+ oscillation, which determines the physiological functions of HMSCs. J. Cell. Physiol. 218: 285–293, 2009.


Journal of Biological Chemistry | 2008

Visualization of Polarized Membrane Type 1 Matrix Metalloproteinase Activity in Live Cells by Fluorescence Resonance Energy Transfer Imaging

Mingxing Ouyang; Shaoying Lu; Xiao Yan Li; Jing Xu; Jihye Seong; Ben N. G. Giepmans; John Y.-J. Shyy; Stephen J. Weiss; Yingxiao Wang

Membrane type 1 matrix metalloproteinase (MT1-MMP) plays a critical role in cancer cell biology by proteolytically remodeling the extracellular matrix. Utilizing fluorescence resonance energy transfer (FRET) imaging, we have developed a novel biosensor, with its sensing element anchoring at the extracellular surface of cell membrane, to visualize MT1-MMP activity dynamically in live cells with subcellular resolution. Epidermal growth factor (EGF) induced significant FRET changes in cancer cells expressing MT1-MMP, but not in MT1-MMP-deficient cells. EGF-induced FRET changes in MT1-MMP-deficient cells could be restored after reconstituting with wild-type MT1-MMP, but not MMP-2, MMP-9, or inactive MT1-MMP mutants. Deletion of the transmembrane domain in the biosensor or treatment with tissue inhibitor of metalloproteinase-2, a cell-impermeable MT1-MMP inhibitor, abolished the EGF-induced FRET response, indicating that MT1-MMP acts at the cell surface to generate FRET changes. In response to EGF, active MT1-MMP was directed to the leading edge of migrating cells along micropatterned fibronectin stripes, in tandem with the local accumulation of the EGF receptor, via a process dependent upon an intact cytoskeletal network. Hence, the MT1-MMP biosensor provides a powerful tool for characterizing the molecular processes underlying the spatiotemporal regulation of this critical class of enzymes.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Distinct biophysical mechanisms of focal adhesion kinase mechanoactivation by different extracellular matrix proteins

Jihye Seong; Arash Tajik; Jie Sun; Jun-Lin Guan; Martin J. Humphries; Susan E. Craig; Asha Shekaran; Andrés J. García; Shaoying Lu; Michael Z. Lin; Ning Wang; Yingxiao Wang

Significance Mechanical forces, which guide cellular functions, can be sensed and translated into biochemical information at focal adhesions, where cells physically connect to extracellular matrix (ECM) through transmembrane receptor integrins. Our results have identified that different ECM proteins, type 1 collagen (Col I) and fibronectin (FN), can either transmit or shield from mechanical forces when regulating crucial intracellular signaling, focal adhesion kinase (FAK), with their different accessibility to the corresponding integrin receptors. Whereas the integrin α2 binding site in Col I is constitutively accessible, mechanical tension is required to expose the integrin α5 binding motif in FN. This finding should advance our understanding on how cells perceive extracellular mechanical cues through natural surface materials. Matrix mechanics controls cell fate by modulating the bonds between integrins and extracellular matrix (ECM) proteins. However, it remains unclear how fibronectin (FN), type 1 collagen, and their receptor integrin subtypes distinctly control force transmission to regulate focal adhesion kinase (FAK) activity, a crucial molecular signal governing cell adhesion/migration. Here we showed, using a genetically encoded FAK biosensor based on fluorescence resonance energy transfer, that FN-mediated FAK activation is dependent on the mechanical tension, which may expose its otherwise hidden FN synergy site to integrin α5. In sharp contrast, the ligation between the constitutively exposed binding motif of type 1 collagen and its receptor integrin α2 was surprisingly tension-independent to induce sufficient FAK activation. Although integrin α subunit determines mechanosensitivity, the ligation between α subunit and the ECM proteins converges at the integrin β1 activation to induce FAK activation. We further discovered that the interaction of the N-terminal protein 4.1/ezrin/redixin/moesin basic patch with phosphatidylinositol 4,5-biphosphate is crucial during cell adhesion to maintain the FAK activation from the inhibitory effect of nearby protein 4.1/ezrin/redixin/moesin acidic sites. Therefore, different ECM proteins either can transmit or can shield from mechanical forces to regulate cellular functions, with the accessibility of ECM binding motifs by their specific integrin α subunits determining the biophysical mechanisms of FAK activation during mechanotransduction.


Nature Communications | 2011

Detection of focal adhesion kinase activation at membrane microdomains by fluorescence resonance energy transfer

Jihye Seong; Mingxing Ouyang; Tae-Jin Kim; Jie Sun; Po-Chao Wen; Shaoying Lu; Yue Zhuo; Nicholas M. Llewellyn; David D. Schlaepfer; Jun-Lin Guan; Shu Chien; Yingxiao Wang

Proper subcellular localization of focal adhesion kinase (FAK) is crucial for many cellular processes. It remains, however, unclear how FAK activity is regulated at subcellular compartments. To visualize the FAK activity at different membrane microdomains, we develop a fluorescence resonance energy transfer (FRET)-based FAK biosensor, and target it into or outside of detergent-resistant membrane (DRM) regions at the plasma membrane. Here we show that, on cell adhesion to extracellular matrix proteins or stimulation by platelet-derived growth factor (PDGF), the FRET responses of DRM-targeting FAK biosensor are stronger than that at non-DRM regions, suggesting that FAK activation can occur at DRM microdomains. Further experiments reveal that the PDGF-induced FAK activation is mediated and maintained by Src activity, whereas FAK activation on cell adhesion is independent of, and in fact essential for the Src activation. Therefore, FAK is activated at membrane microdomains with distinct activation mechanisms in response to different physiological stimuli.


Chemistry & Biology | 2009

Visualization of Src Activity at Different Compartments of the Plasma Membrane by FRET Imaging

Jihye Seong; Shaoying Lu; Mingxing Ouyang; He Huang; Jin Zhang; Margaret C. Frame; Yingxiao Wang

Membrane compartments function as segregated signaling platforms for different cellular functions. It is not clear how Src is regulated at different membrane compartments. To visualize local Src activity in live cells, a FRET-based Src biosensor was targeted in or outside of lipid rafts at the plasma membrane, via acylation or prenylation modifications on targeting tags either directly fused to the biosensor or coupled to the biosensor through an inducible heterodimerization system. In response to growth factors and pervanadate, the induction of Src activity in rafts was slower and weaker, dependent on actin and possibly its mediated transportation of Src from perinuclear regions to the plasma membrane. In contrast, the induction of Src activity in nonrafts was faster and stronger, dependent on microtubules. Hence, Src activity is differentially regulated via cytoskeleton at different membrane compartments.


PLOS Computational Biology | 2008

The Spatiotemporal Pattern of Src Activation at Lipid Rafts Revealed by Diffusion-Corrected FRET Imaging

Shaoying Lu; Mingxing Ouyang; Jihye Seong; Jin Zhang; Shu Chien; Yingxiao Wang

Genetically encoded biosensors based on fluorescence resonance energy transfer (FRET) have been widely applied to visualize the molecular activity in live cells with high spatiotemporal resolution. However, the rapid diffusion of biosensor proteins hinders a precise reconstruction of the actual molecular activation map. Based on fluorescence recovery after photobleaching (FRAP) experiments, we have developed a finite element (FE) method to analyze, simulate, and subtract the diffusion effect of mobile biosensors. This method has been applied to analyze the mobility of Src FRET biosensors engineered to reside at different subcompartments in live cells. The results indicate that the Src biosensor located in the cytoplasm moves 4–8 folds faster (0.93±0.06 µm2/sec) than those anchored on different compartments in plasma membrane (at lipid raft: 0.11±0.01 µm2/sec and outside: 0.18±0.02 µm2/sec). The mobility of biosensor at lipid rafts is slower than that outside of lipid rafts and is dominated by two-dimensional diffusion. When this diffusion effect was subtracted from the FRET ratio images, high Src activity at lipid rafts was observed at clustered regions proximal to the cell periphery, which remained relatively stationary upon epidermal growth factor (EGF) stimulation. This result suggests that EGF induced a Src activation at lipid rafts with well-coordinated spatiotemporal patterns. Our FE-based method also provides an integrated platform of image analysis for studying molecular mobility and reconstructing the spatiotemporal activation maps of signaling molecules in live cells.


Nature Communications | 2013

N-cadherin regulates spatially polarized signals through distinct p120ctn and β-catenin-dependent signalling pathways

Mingxing Ouyang; Shaoying Lu; Tae-Jin Kim; Chin En Chen; Jihye Seong; Deborah E. Leckband; Fei Wang; Albert B. Reynolds; Martin A. Schwartz; Yingxiao Wang

The spatial distribution of molecular signals within cells is crucial for cellular functions. Here, as a model to study the polarized spatial distribution of molecular activities, we used cells on micro-patterned strips of fibronectin with one end free and the other end contacting a neighboring cell. Phosphoinositide 3-kinase (PI3K) and the small GTPase Rac display greater activity at the free end, whereas myosin II light chain (MLC) and actin filaments are enriched near the intercellular junction. PI3K and Rac polarization depend specifically on the N-cadherin-p120ctn complex, whereas MLC and actin filament polarization depend on the N-cadherin-β-catenin complex. Integrins promote high PI3K/Rac activities at the free end, and the N-cadherin–p120ctn complex excludes integrin α5 at the junctions to suppress local PI3K and Rac activity. We hence conclude that N-cadherin couples with distinct effectors to polarize PI3K/Rac and MLC/actin filaments in migrating cells.


Nano Letters | 2015

Activatable and Cell-Penetrable Multiplex FRET Nanosensor for Profiling MT1-MMP Activity in Single Cancer Cells

Eddie Y. Chung; Christopher J. Ochs; Yi Wang; Lei Lei; Qin Qin; Andrew M. Smith; Alex Y. Strongin; Roger D. Kamm; Ying Xin Qi; Shaoying Lu; Yingxiao Wang

We developed a quantum-dot-based fluorescence resonance energy transfer (QD-FRET) nanosensor to visualize the activity of matrix metalloproteinase (MT1-MMP) at cell membrane. A bended peptide with multiple motifs was engineered to position the FRET pair at a close proximity to allow energy transfer, which can be cleaved by active MT1-MMP to result in FRET changes and the exposure of cell penetrating sequence. Via FRET and penetrated QD signals, the nanosensor can profile cancer cells.


PLOS ONE | 2013

Quantitative FRET imaging to visualize the invasiveness of live breast cancer cells.

Shaoying Lu; Yi Wang; He Huang; Yijia Pan; Eric J. Chaney; Stephen A. Boppart; Howard Ozer; Alex Y. Strongin; Yingxiao Wang

Matrix metalloproteinases (MMPs) remodel tumor microenvironment and promote cancer metastasis. Among the MMP family proteases, the proteolytic activity of the pro-tumorigenic and pro-metastatic membrane-type 1 (MT1)-MMP constitutes a promising and targetable biomarker of aggressive cancer tumors. In this study, we systematically developed and characterized several highly sensitive and specific biosensors based on fluorescence resonant energy transfer (FRET), for visualizing MT1-MMP activity in live cells. The sensitivity of the AHLR-MT1-MMP biosensor was the highest and five times that of a reported version. Hence, the AHLR biosensor was employed to quantitatively profile the MT1-MMP activity in multiple breast cancer cell lines, and to visualize the spatiotemporal MT1-MMP activity simultaneously with the underlying collagen matrix at the single cell level. We detected a significantly higher level of MT1-MMP activity in invasive cancer cells than those in benign or non-invasive cells. Our results further show that the high MT1-MMP activity was stimulated by the adhesion of invasive cancer cells onto the extracellular matrix, which is precisely correlated with the cell’s ability to degrade the collagen matrix. Thus, we systematically optimized a FRET-based biosensor, which provides a powerful tool to detect the pro-invasive MT1-MMP activity at single cell levels. This readout can be applied to profile the invasiveness of single cells from clinical samples, and to serve as an indicator for screening anti-cancer inhibitors.


PLOS ONE | 2011

Computational analysis of the spatiotemporal coordination of polarized PI3K and Rac1 activities in micro-patterned live cells.

Shaoying Lu; Tae-Jin Kim; Chih-En Chen; Mingxing Ouyang; Jihye Seong; Xiaoling Liao; Yingxiao Wang

Polarized molecular activities play important roles in guiding the cell toward persistent and directional migration. In this study, the polarized distributions of the activities of phosphatidylinositol 3-kinase (PI3K) and the Rac1 small GTPase were monitored using chimeric fluorescent proteins (FPs) in cells constrained on micro-patterned strips, with one end connecting to a neighboring cell (junction end) and the other end free of cell-cell contact (free end). The recorded spatiotemporal dynamics of the fluorescent intensity from different cells was scaled into a uniform coordinate system and applied to compute the molecular activity landscapes in space and time. The results revealed different polarization patterns of PI3K and Rac1 activity induced by the growth factor stimulation. The maximal intensity of different FPs, and the edge position and velocity at the free end were further quantified to analyze their correlation and decipher the underlying signaling sequence. The results suggest that the initiation of the edge extension occurred before the activation of PI3K, which led to a stable extension of the free end followed by the Rac1 activation. Therefore, the results support a concerted coordination of sequential signaling events and edge dynamics, underscoring the important roles played by PI3K activity at the free end in regulating the stable lamellipodia extension and cell migration. Meanwhile, the quantification methods and accompanying software developed can provide a convenient and powerful computational analysis platform for the study of spatiotemporal molecular distribution and hierarchy in live cells based on fluorescence images.

Collaboration


Dive into the Shaoying Lu's collaboration.

Top Co-Authors

Avatar

Yingxiao Wang

University of California

View shared research outputs
Top Co-Authors

Avatar

Shu Chien

University of California

View shared research outputs
Top Co-Authors

Avatar

Xiaoling Liao

Chongqing University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Jin Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar

Jun-Lin Guan

University of Cincinnati Academic Health Center

View shared research outputs
Top Co-Authors

Avatar

Wenfeng Xu

Chongqing University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrés J. García

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Asha Shekaran

Georgia Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge