Network


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

Hotspot


Dive into the research topics where Yuanjian Guo is active.

Publication


Featured researches published by Yuanjian Guo.


FEBS Letters | 2002

Multiple roles of pleckstrin homology domains in phospholipase Cβ function

Finly Philip; Yuanjian Guo; Suzanne Scarlata

Since their discovery almost 10 years ago pleckstrin homology (PH) domains have been identified in a wide variety of proteins. Here, we focus on two proteins whose PH domains play a defined functional role, phospholipase C (PLC)‐β2 and PLCδ1. While the PH domains of both proteins are responsible for membrane targeting, their specificity of membrane binding drastically differs. However, in both these proteins the PH domains work to modulate the activity of their catalytic core upon interaction with either phosphoinositol lipids or G protein activators. These observations show that these PH domains are not simply binding sites tethered onto their host enzyme but are intimately associated with their catalytic core. This property may be true for other PH domains.


Journal of Biological Chemistry | 2005

Phospholipase Cbeta2 binds to and inhibits phospholipase Cdelta1.

Yuanjian Guo; Mario Rebecchi; Suzanne Scarlata

Phospholipase Cβ (PLCβ) isoforms, which are under the control of Gαq and Gβγ subunits, generate Ca2+ signals induced by a broad array of extracellular agonists, whereas PLCδ isoforms depend on a rise in cytosolic Ca2+ for their activation. Here we find that PLCβ2 binds strongly to PLCδ1 and inhibits its catalytic activity in vitro and in living cells. In vitro, this PLC complex can be disrupted by increasing concentrations of free Gβγ subunits. Such competition has consequences for signaling, because in HEK293 cells PLCβ2 suppresses elevated basal [Ca2+] and inositol phosphates levels and the sustained agonist-induced elevation of Ca2+ levels caused by PLCδ1. Also, expression of both PLCs results in a synergistic release of [Ca2+] upon stimulation in A10 cells. These results support a model in which PLCβ2 suppresses the basal catalytic activity of PLCδ1, which is relieved by binding of Gβγ subunits to PLCβ2 allowing for amplified calcium signals.


Cellular Signalling | 2012

α-Synuclein increases the cellular level of phospholipase Cβ1.

Yuanjian Guo; Barbara Rosati; Suzanne Scarlata

α-Synuclein is a conserved protein that is a key component in neurodegenerative plaques [1,2]. α-Synuclein binds strongly to phospholipase Cβ (PLCβ) and promotes Ca2+ release in cells. Here, we show that expression of α-synuclein increases the cellular level of PLCβ1 in two neuronal cell lines: PC12 and SK-N-S-SH. The increase in PLCβ1 is not accompanied by changes in the level of RNA or in ubiquitination. Instead, we find that α-synuclein protects PLCβ1 from trypsin digestion and from degradation by the Ca(+2) activated protease calpain. Calpain removes the C-terminal region of the enzyme which mediates activation by Gα(q). We find that in SK-N-SH cells, α-synuclein reduced degradation of PLCβ1 by calpain during Ca2+ signaling allowing the enzyme to remain sensitive to Gα(q) activation. Taken together, our studies show that α-synuclein protects the integrity of PLCβ1 and its ability to be activated by Gα(q), which may in turn impact Ca2+ signaling.


The FASEB Journal | 2012

Phospholipase Cβ1 is linked to RNA interference of specific genes through translin-associated factor X

Finly Philip; Yuanjian Guo; Omoz Aisiku; Suzanne Scarlata

Phospholipase Cβ1 (PLCβ1) is a G‐protein‐regulated enzyme whose activity results in proliferative and mitogenic changes in the cell. We have previously found that in solution PLCβ1 binds to the RNA processing protein translin‐associated factor X (TRAX) with nanomolar affinity and that this binding competes with G proteins. Here, we show that endogenous PLCβ1 and TRAX interact in SK‐N‐SH cells and also in HEK293 cells induced to overexpress PLCβ1. In HEK293 cells, TRAX overexpression ablates Ca2+ signals generated by G protein‐PLCβ1 activation. TRAX plays a key role in down‐regulation of proteins by small, interfering RNA, and PLCβ1 overexpression completely reverses the 2‐ to 4‐fold down‐regulation of GAPDH by siRNA in HEK293 and HeLa cells as seen by an ~4‐fold recovery in both the transcript and protein levels. Also, down‐regulation of endogenous PLCβ1 in HEK293 and HeLa cells allows for an ~20% increase in siRNA(GAPDH) silencing. While PLCβ1 overexpression results in a 50% reversal of cell death caused by siRNA(LDH), it does not affect cell survival or silencing of other genes (e.g., cyclophilin, Hsp90, translin). PLCβ1 overexpression in HEK293 and HeLa cells causes a 30% reduction in the total amount of small RNAs. LDH and GAPDH are part of a complex that promotes H2B synthesis that allows cells to progress through the S phase. We find that PLCβ1 reverses the cell death and completely rescues H2B levels caused by siRNA knockdown of LDH or GAPDH. Taken together, our study shows a novel role of PLCβ1 in gene regulation through TRAX association.—Philip, F., Guo, Y., Aisiku, O., Scarlata, S. Phospholipase Cβ1 is linked to RNA interference of specific genes through translin‐associated factor X. FASEB J. 26, 4903–4913 (2012). www.fasebj.org


Advances in biological regulation | 2016

Phospholipase Cβ connects G protein signaling with RNA interference.

Suzanne Scarlata; Osama Garwain; Leo Williams; Imanol Gonzalez Burguera; Barbara Rosati; Shriya Sahu; Yuanjian Guo; Finly Philip; Urszula Golebiewska

Phosphoinositide-specific-phospholipase Cβ (PLCβ) is the main effector of Gαq stimulation which is coupled to receptors that bind acetylcholine, bradykinin, dopamine, angiotensin II as well as other hormones and neurotransmitters. Using a yeast two-hybrid and other approaches, we have recently found that the same region of PLCβ that binds Gαq also interacts with Component 3 Promoter of RNA induced silencing complex (C3PO), which is required for efficient activity of the RNA-induced silencing complex. In purified form, C3PO competes with Gαq for PLCβ binding and at high concentrations can quench PLCβ activation. Additionally, we have found that the binding of PLCβ to C3PO inhibits its nuclease activity leading to reversal of RNA-induced silencing of specific genes. In cells, we found that PLCβ distributes between the plasma membrane where it localizes with Gαq, and in the cytosol where it localizes with C3PO. When cells are actively processing small interfering RNAs the interaction between PLCβ and C3PO gets stronger and leads to changes in the cellular distribution of PLCβ. The magnitude of attenuation is specific for different silencing RNAs. Our studies imply a direct link between calcium responses mediated through Gαq and post-transcriptional gene regulation through PLCβ.


Journal of Biological Chemistry | 2010

The Small G Protein Rac1 Activates Phospholipase Cδ1 through Phospholipase Cβ2

Yuanjian Guo; Urszula Golebiewska; Stephen D'Amico; Suzanne Scarlata

Rac1, which is associated with cytoskeletal pathways, can activate phospholipase Cβ2 (PLCβ2) to increase intracellular Ca2+ levels. This increased Ca2+ can in turn activate the very robust PLCδ1 to synergize Ca2+ signals. We have previously found that PLCβ2 will bind to and inhibit PLCδ1 in solution by an unknown mechanism and that PLCβ2·PLCδ1 complexes can be disrupted by Gβγ subunits. However, because the major populations of PLCβ2 and PLCδ1 are cytosolic, their regulation by Gβγ subunits is not clear. Here, we have found that the pleckstrin homology (PH) domains of PLCβ2 and PLCβ3 are the regions that result in PLCδ1 binding and inhibition. In cells, PLCβ2·PLCδ1 form complexes as seen by Förster resonance energy transfer and co-immunoprecipitation, and microinjection of PHβ2 dissociates the complex. Using PHβ2 as a tool to assess the contribution of PLCβ inhibition of PLCδ1 to Ca2+ release, we found that, although PHβ2 only results in a 25% inhibition of PLCδ1 in solution, in cells the presence of PHβ2 appears to eliminates Ca2+ release suggesting a large threshold effect. We found that the small plasma membrane population of PLCβ2·PLCδ1 is disrupted by activation of heterotrimeric G proteins, and that the major cytosolic population of the complexes are disrupted by Rac1 activation. Thus, the activity of PLCδ1 is controlled by the amount of bound PLCβ2 that changes with displacement of the enzyme by heterotrimeric or small G proteins. Through PLCβ2, PLCδ1 activation is linked to surface receptors as well as signals that mediate cytoskeletal pathways.


Biophysical Journal | 2011

Modulation of Ca2+ Activity in Cardiomyocytes through Caveolae-Gαq Interactions

Yuanjian Guo; Urszula Golebiewska; Suzanne Scarlata

Cardiomyocytes have a complex Ca(2+) behavior and changes in this behavior may underlie certain disease states. Intracellular Ca(2+) activity can be regulated by the phospholipase Cβ-Gα(q) pathway localized on the plasma membrane. The plasma membranes of cardiomycoytes are rich in caveolae domains organized by caveolin proteins. Caveolae may indirectly affect cell signals by entrapping and localizing specific proteins. Recently, we found that caveolin may specifically interact with activated Gα(q), which could affect Ca(2+) signals. Here, using fluorescence imaging and correlation techniques we show that Gα(q)-Gβγ subunits localize to caveolae in adult ventricular canine cardiomyoctyes. Carbachol stimulation releases Gβγ subunits from caveolae with a concurrent stabilization of activated Gα(q) by caveolin-3 (Cav3). These cells show oscillating Ca(2+) waves that are not seen in neonatal cells that do not contain Cav3. Microinjection of a peptide that disrupts Cav3-Gα(q) association, but not a control peptide, extinguishes the waves. Furthermore, these waves are unchanged with rynaodine treatment, but not seen with treatment of a phospholipase C inhibitor, implying that Cav3-Gα(q) is responsible for this Ca(2+) activity. Taken together, these studies show that caveolae play a direct and active role in regulating basal Ca(2+) activity in cardiomyocytes.


Journal of Biological Chemistry | 2009

Evidence for a Second, High Affinity Gβγ Binding Site on Gαi1(GDP) Subunits

Jingting Wang; Parijat Sengupta; Yuanjian Guo; Urszula Golebiewska; Suzanne Scarlata

It is well known that Gαi1(GDP) binds strongly to Gβγ subunits to form the Gαi1(GDP)-Gβγ heterotrimer, and that activation to Gαi1(GTP) results in conformational changes that reduces its affinity for Gβγ subunits. Previous studies of G protein subunit interactions have used stoichiometric amounts of the proteins. Here, we have found that Gαi1(GDP) can bind a second Gβγ subunit with an affinity only 10-fold weaker than the primary site and close to the affinity between activated Gαi1 and Gβγ subunits. Also, we find that phospholipase Cβ2, an effector of Gβγ, does not compete with the second binding site implying that effectors can be bound to the Gαi1(GDP)-(Gβγ)2 complex. Biophysical measurements and molecular docking studies suggest that this second site is distant from the primary one. A synthetic peptide having a sequence identical to the putative second binding site on Gαi1 competes with binding of the second Gβγ subunit. Injection of this peptide into cultured cells expressing eYFP-Gαi1(GDP) and eCFP-Gβγ reduces the overall association of the subunits suggesting this site is operative in cells. We propose that this second binding site serves to promote and stabilize G protein subunit interactions in the presence of competing cellular proteins.


Biochemistry | 2013

A Loss in Cellular Protein Partners Promotes α-Synuclein Aggregation in Cells Resulting from Oxidative Stress

Yuanjian Guo; Suzanne Scarlata

There is a consensus that oxidative stress promotes neurodegeneration and may be linked to plaque formation. α-Synuclein is the main component of neurodegenerative plaques. We have found that α-synuclein binds strongly to the enzyme phospholipase Cβ1 (PLCβ1) in vitro and in cells affecting both its G protein activation and its degradation. Because PLCβ1 binds to α-synuclein in cells, we tested whether decreasing its level would promote α-synuclein aggregation and whether overproducing PLCβ1 would inhibit aggregation. By imaging fluorescent α-synuclein in living HEK293, PC12, and SK-H-SH cells, we find that α-synuclein aggregation is directly related to the level of PLCβ1. Importantly, we found that oxidative stress does not affect the cellular levels of α-synuclein but results in the down-regulation of PLCβ1 thereby promoting α-synuclein aggregation. A peptide that mimics part of the α-synuclein binding site to PLCβ prevents aggregation. Our studies indicate that PLCβ1 can reduce cell damage under oxidative stress and offers a potential site that might be exploited to prevent α-synuclein aggregation.


Journal of Cardiovascular Pharmacology | 2012

IMPROvING CARDIAC CONDuCTION wITH A SkElETAl MuSClE SODIuM CHANNEl By GENE AND CEll THERAPy

Jia Lu; Hong-Zhan Wang; Zhiheng Jia; Joan Zuckerman; Zhongju Lu; Yuanjian Guo; Gerard J.J. Boink; Peter R. Brink; Richard B. Robinson; Emilia Entcheva; Ira S. Cohen

Abstract: The voltage-gated Na+ channel is a critical determinant of the action potential (AP) upstroke. Increasing Na+ conductance may speed AP propagation. In this study, we propose use of the skeletal muscle Na+ channel SkM1 as a more favorable gene than the cardiac isoform SCN5A to enhance conduction velocity in depolarized cardiac tissue. We used cells that electrically coupled with cardiac myocytes as a delivery platform to introduce the Na+ channels. Human embryonic kidney 293 cells were stably transfected with SkM1 or SCN5A. SkM1 had a more depolarized (18 mV shift) inactivation curve than SCN5A. We also found that SkM1 recovered faster from inactivation than SCN5A. When coupled with SkM1 expressing cells, cultured myocytes showed an increase in the dV/dtmax of the AP. Expression of SCN5A had no such effect. In an in vitro cardiac syncytium, coculture of neonatal cardiac myocytes with SkM1 expressing but not SCN5A expressing cells significantly increased the conduction velocity under both normal and depolarized conditions. In an in vitro reentry model induced by high-frequency stimulation, expression of SkM1 also enhanced angular velocity of the induced reentry. These results suggest that cells carrying a Na+ channel with a more depolarized inactivation curve can improve cardiac excitability and conduction in depolarized tissues.

Collaboration


Dive into the Yuanjian Guo's collaboration.

Top Co-Authors

Avatar

Suzanne Scarlata

Worcester Polytechnic Institute

View shared research outputs
Top Co-Authors

Avatar

Urszula Golebiewska

Queensborough Community College

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
Top Co-Authors

Avatar

Jia Lu

Stony Brook University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge