Network


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

Hotspot


Dive into the research topics where Qiuyan Chen is active.

Publication


Featured researches published by Qiuyan Chen.


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

Involvement of distinct arrestin-1 elements in binding to different functional forms of rhodopsin

Tiandi Zhuang; Qiuyan Chen; Min-Kyu Cho; Sergey A. Vishnivetskiy; Tina M. Iverson; Vsevolod V. Gurevich; Charles R. Sanders

Solution NMR spectroscopy of labeled arrestin-1 was used to explore its interactions with dark-state phosphorylated rhodopsin (P-Rh), phosphorylated opsin (P-opsin), unphosphorylated light-activated rhodopsin (Rh*), and phosphorylated light-activated rhodopsin (P-Rh*). Distinct sets of arrestin-1 elements were seen to be engaged by Rh* and inactive P-Rh, which induced conformational changes that differed from those triggered by binding of P-Rh*. Although arrestin-1 affinity for Rh* was seen to be low (KD > 150 μM), its affinity for P-Rh (KD ∼80 μM) was comparable to the concentration of active monomeric arrestin-1 in the outer segment, suggesting that P-Rh generated by high-gain phosphorylation is occupied by arrestin-1 under physiological conditions and will not signal upon photo-activation. Arrestin-1 was seen to bind P-Rh* and P-opsin with fairly high affinity (KD of ∼50 and 800 nM, respectively), implying that arrestin-1 dissociation is triggered only upon P-opsin regeneration with 11-cis-retinal, precluding noise generated by opsin activity. Based on their observed affinity for arrestin-1, P-opsin and inactive P-Rh very likely affect the physiological monomer-dimer-tetramer equilibrium of arrestin-1, and should therefore be taken into account when modeling photoreceptor function. The data also suggested that complex formation with either P-Rh* or P-opsin results in a global transition in the conformation of arrestin-1, possibly to a dynamic molten globule-like structure. We hypothesize that this transition contributes to the mechanism that triggers preferential interactions of several signaling proteins with receptor-activated arrestins.


Journal of Biological Chemistry | 2013

Engineering Visual Arrestin-1 with Special Functional Characteristics

Sergey A. Vishnivetskiy; Qiuyan Chen; Maria C. Palazzo; Evan K. Brooks; Christian Altenbach; Tina M. Iverson; Wayne L. Hubbell; Vsevolod V. Gurevich

Background: Arrestin-1 with enhanced binding to unphosphorylated active rhodopsin (Rh*) has therapeutic potential. Results: Manipulation of the rhodopsin binding surface of arrestin-1 greatly increases its binding to Rh*. Conclusion: Stable arrestin-1 with high binding to Rh* can be engineered with and without the ability to self-associate. Significance: The affinity of arrestin-1 for Rh* and its propensity to oligomerize can be independently changed by targeted mutagenesis. Arrestin-1 preferentially binds active phosphorylated rhodopsin. Previously, a mutant with enhanced binding to unphosphorylated active rhodopsin (Rh*) was shown to partially compensate for lack of rhodopsin phosphorylation in vivo. Here we showed that reengineering of the receptor binding surface of arrestin-1 further improves the binding to Rh* while preserving protein stability. In mammals, arrestin-1 readily self-associates at physiological concentrations. The biological role of this phenomenon can only be elucidated by replacing wild type arrestin-1 in living animals with a non-oligomerizing mutant retaining all other functions. We demonstrate that constitutively monomeric forms of arrestin-1 are sufficiently stable for in vivo expression. We also tested the idea that individual functions of arrestin-1 can be independently manipulated to generate mutants with the desired combinations of functional characteristics. Here we showed that this approach is feasible; stable forms of arrestin-1 with high Rh* binding can be generated with or without the ability to self-associate. These novel molecular tools open the possibility of testing of the biological role of arrestin-1 self-association and pave the way to elucidation of full potential of compensational approach to gene therapy of gain-of-function receptor mutations.


Scientific Reports | 2016

Peptide mini-scaffold facilitates JNK3 activation in cells

Xuanzhi Zhan; Henriette Stoy; Tamer S. Kaoud; Nicole A. Perry; Qiuyan Chen; Alejandro Perez; Sylvia Els-Heindl; Jack V. Slagis; Tina M. Iverson; Annette G. Beck-Sickinger; Eugenia V. Gurevich; Kevin N. Dalby; Vsevolod V. Gurevich

Three-kinase mitogen-activated protein kinase (MAPK) signaling cascades are present in virtually all eukaryotic cells. MAPK cascades are organized by scaffold proteins, which assemble cognate kinases into productive signaling complexes. Arrestin-3 facilitates JNK activation in cells, and a short 25-residue arrestin-3 peptide was identified as the critical JNK3-binding element. Here we demonstrate that this peptide also binds MKK4, MKK7, and ASK1, which are upstream JNK3-activating kinases. This peptide is sufficient to enhance JNK3 activity in cells. A homologous arrestin-2 peptide, which differs only in four positions, binds MKK4, but not MKK7 or JNK3, and is ineffective in cells at enhancing activation of JNK3. The arrestin-3 peptide is the smallest MAPK scaffold known. This peptide or its mimics can regulate MAPKs, affecting cellular decisions to live or die.


Cellular Signalling | 2013

Rapid degeneration of rod photoreceptors expressing self-association-deficient arrestin-1 mutant.

Xiufeng Song; Jungwon Seo; Faiza Baameur; Sergey A. Vishnivetskiy; Qiuyan Chen; Seunghyi Kook; Miyeon Kim; Evan K. Brooks; Christian Altenbach; Yuan Hong; Susan M. Hanson; Maria C. Palazzo; Jeannie Chen; Wayne L. Hubbell; Eugenia V. Gurevich; Vsevolod V. Gurevich

Arrestin-1 binds light-activated phosphorhodopsin and ensures timely signal shutoff. We show that high transgenic expression of an arrestin-1 mutant with enhanced rhodopsin binding and impaired oligomerization causes apoptotic rod death in mice. Dark rearing does not prevent mutant-induced cell death, ruling out the role of arrestin complexes with light-activated rhodopsin. Similar expression of WT arrestin-1 that robustly oligomerizes, which leads to only modest increase in the monomer concentration, does not affect rod survival. Moreover, WT arrestin-1 co-expressed with the mutant delays retinal degeneration. Thus, arrestin-1 mutant directly affects cell survival via binding partner(s) other than light-activated rhodopsin. Due to impaired self-association of the mutant its high expression dramatically increases the concentration of the monomer. The data suggest that monomeric arrestin-1 is cytotoxic and WT arrestin-1 protects rods by forming mixed oligomers with the mutant and/or competing with it for the binding to non-receptor partners. Thus, arrestin-1 self-association likely serves to keep low concentration of the toxic monomer. The reduction of the concentration of harmful monomer is an earlier unappreciated biological function of protein oligomerization.


Nature Communications | 2017

Structural basis of arrestin-3 activation and signaling

Qiuyan Chen; Nicole A. Perry; Sergey A. Vishnivetskiy; Sandra Berndt; Nathaniel C. Gilbert; Ya Zhuo; Prashant K. Singh; Jonas Tholen; Melanie D. Ohi; Eugenia V. Gurevich; Chad A. Brautigam; Candice S. Klug; Vsevolod V. Gurevich; Tina M. Iverson

A unique aspect of arrestin-3 is its ability to support both receptor-dependent and receptor-independent signaling. Here, we show that inositol hexakisphosphate (IP6) is a non-receptor activator of arrestin-3 and report the structure of IP6-activated arrestin-3 at 2.4-Å resolution. IP6-activated arrestin-3 exhibits an inter-domain twist and a displaced C-tail, hallmarks of active arrestin. IP6 binds to the arrestin phosphate sensor, and is stabilized by trimerization. Analysis of the trimerization surface, which is also the receptor-binding surface, suggests a feature called the finger loop as a key region of the activation sensor. We show that finger loop helicity and flexibility may underlie coupling to hundreds of diverse receptors and also promote arrestin-3 activation by IP6. Importantly, we show that effector-binding sites on arrestins have distinct conformations in the basal and activated states, acting as switch regions. These switch regions may work with the inter-domain twist to initiate and direct arrestin-mediated signaling.While arrestins are mainly associated with GPCR signaling, arrestin-3 can signal independently of receptor interaction. Here the authors present the structure of arrestin-3 bound to inositol hexakisphosphate (IP6) and propose a model for arrestin-3 activation.


Current protocols in pharmacology | 2014

Arrestin expression in E. coli and purification.

Sergey A. Vishnivetskiy; Xuanzhi Zhan; Qiuyan Chen; Tina M. Iverson; Vsevolod V. Gurevich

Purified arrestin proteins are necessary for biochemical, biophysical, and crystallographic studies of these versatile regulators of cell signaling. Described herein is a basic protocol for arrestin expression in E. coli and purification of the tag‐free wild‐type and mutant arrestins. The method includes ammonium sulfate precipitation of arrestins from cell lysates, followed by heparin‐Sepharose chromatography. Depending on the arrestin type and/or mutations, the next step is Q‐Sepharose or SP‐Sepharose chromatography. In many cases the nonbinding column is used as a filter to bind contaminants without retaining arrestin. In some cases both chromatographic steps must be performed sequentially to achieve high purity. Purified arrestins can be concentrated up to 10 mg/ml, remain fully functional, and withstand several cycles of freezing and thawing, provided that overall salt concentration is maintained at or above physiological levels.


Trends in Biochemical Sciences | 2018

Structural Basis of Arrestin-Dependent Signal Transduction

Qiuyan Chen; Tina M. Iverson; Vsevolod V. Gurevich

Arrestins are a small family of proteins with four isoforms in humans. Remarkably, two arrestins regulate signaling from >800 G protein-coupled receptors (GPCRs) or nonreceptor activators by simultaneously binding an activator and one out of hundreds of other signaling proteins. When arrestins are bound to GPCRs or other activators, the affinity for these signaling partners changes. Thus, it is proposed that an activator alters arrestins ability to transduce a signal. The comparison of all available arrestin structures identifies several common conformational rearrangements associated with activation. In particular, it identifies elements that are directly involved in binding to GPCRs or other activators, elements that likely engage distinct downstream effectors, and elements that likely link the activator-binding sites with the effector-binding sites.


Analytical Biochemistry | 2018

Using two-site binding models to analyze microscale thermophoresis data

Shih Chia Tso; Qiuyan Chen; Sergey A. Vishnivetskiy; Vsevolod V. Gurevich; Tina M. Iverson; Chad A. Brautigam

The emergence of microscale thermophoresis (MST) as a technique for determining the dissociation constants for bimolecular interactions has enabled these quantities to be measured in systems that were previously difficult or impracticable. However, most models for analyses of these data featured the assumption of a simple 1:1 binding interaction. The only model widely used for multiple binding sites was the Hill equation. Here, we describe two new MST analytic models that assume a 1:2 binding scheme: the first features two microscopic binding constants (KD(1) and KD(2)), while the other assumes symmetry in the bivalent molecule, culminating in a model with a single macroscopic dissociation constant (KD,M) and a single factor (α) that accounts for apparent cooperativity in the binding. We also discuss the general applicability of the Hill equation for MST data. The performances of the algorithms on both real and simulated data are assessed, and implementation of the algorithms in the MST analysis program PALMIST is discussed.


Methods of Molecular Biology | 2015

The Rhodopsin-Arrestin-1 Interaction in Bicelles

Qiuyan Chen; Sergey A. Vishnivetskiy; Tiandi Zhuang; Min-Kyu Cho; Tarjani M. Thaker; Charles R. Sanders; Vsevolod V. Gurevich; Tina M. Iverson

G-protein-coupled receptors (GPCRs) are essential mediators of information transfer in eukaryotic cells. Interactions between GPCRs and their binding partners modulate the signaling process. For example, the interaction between GPCR and cognate G protein initiates the signal, while the interaction with cognate arrestin terminates G-protein-mediated signaling. In visual signal transduction, arrestin-1 selectively binds to the phosphorylated light-activated GPCR rhodopsin to terminate rhodopsin signaling. Under physiological conditions, the rhodopsin-arrestin-1 interaction occurs in highly specialized disk membrane in which rhodopsin resides. This membrane is replaced with mimetics when working with purified proteins. While detergents are commonly used as membrane mimetics, most detergents denature arrestin-1, preventing biochemical studies of this interaction. In contrast, bicelles provide a suitable alternative medium. An advantage of bicelles is that they contain lipids, which have been shown to be necessary for normal rhodopsin-arrestin-1 interaction. Here we describe how to reconstitute rhodopsin into bicelles, and how bicelle properties affect the rhodopsin-arrestin-1 interaction.


Handbook of experimental pharmacology | 2014

Self-association of arrestin family members.

Qiuyan Chen; Ya Zhuo; Miyeon Kim; Susan M. Hanson; Derek J. Francis; Sergey A. Vishnivetskiy; Christian Altenbach; Candice S. Klug; Wayne L. Hubbell; Vsevolod V. Gurevich

Collaboration


Dive into the Qiuyan Chen's collaboration.

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
Top Co-Authors

Avatar

Evan K. Brooks

University of California

View shared research outputs
Top Co-Authors

Avatar

Miyeon Kim

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge