Network


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

Hotspot


Dive into the research topics where Sergey A. Vishnivetskiy is active.

Publication


Featured researches published by Sergey A. Vishnivetskiy.


Nature | 2015

Crystal structure of rhodopsin bound to arrestin by femtosecond X-ray laser

Yanyong Kang; X. Edward Zhou; Xiang Gao; Yuanzheng He; Wei Liu; Andrii Ishchenko; Anton Barty; Thomas A. White; Oleksandr Yefanov; Gye Won Han; Qingping Xu; Parker W. de Waal; Jiyuan Ke; M. H.Eileen Tan; Chenghai Zhang; Arne Moeller; Graham M. West; Bruce D. Pascal; Ned Van Eps; Lydia N. Caro; Sergey A. Vishnivetskiy; Regina J. Lee; Kelly Suino-Powell; Xin Gu; Kuntal Pal; Jinming Ma; Xiaoyong Zhi; Sébastien Boutet; Garth J. Williams; Marc Messerschmidt

G-protein-coupled receptors (GPCRs) signal primarily through G proteins or arrestins. Arrestin binding to GPCRs blocks G protein interaction and redirects signalling to numerous G-protein-independent pathways. Here we report the crystal structure of a constitutively active form of human rhodopsin bound to a pre-activated form of the mouse visual arrestin, determined by serial femtosecond X-ray laser crystallography. Together with extensive biochemical and mutagenesis data, the structure reveals an overall architecture of the rhodopsin–arrestin assembly in which rhodopsin uses distinct structural elements, including transmembrane helix 7 and helix 8, to recruit arrestin. Correspondingly, arrestin adopts the pre-activated conformation, with a ∼20° rotation between the amino and carboxy domains, which opens up a cleft in arrestin to accommodate a short helix formed by the second intracellular loop of rhodopsin. This structure provides a basis for understanding GPCR-mediated arrestin-biased signalling and demonstrates the power of X-ray lasers for advancing the frontiers of structural biology.


Structure | 2001

Crystal structure of beta-arrestin at 1.9 A: possible mechanism of receptor binding and membrane Translocation.

May Han; Vsevolod V. Gurevich; Sergey A. Vishnivetskiy; Paul B. Sigler; Carsten Schubert

BACKGROUNDnArrestins are responsible for the desensitization of many sequence-divergent G protein-coupled receptors. They compete with G proteins for binding to activated phosphorylated receptors, initiate receptor internalization, and activate additional signaling pathways.nnnRESULTSnIn order to understand the structural basis for receptor binding and arrestins function as an adaptor molecule, we determined the X-ray crystal structure of two truncated forms of bovine beta-arrestin in its cytosolic inactive state to 1.9 A. Mutational analysis and chimera studies identify the regions in beta-arrestin responsible for receptor binding specificity. beta-arrestin demonstrates high structural homology with the previously solved visual arrestin. All key structural elements responsible for arrestins mechanism of activation are conserved.nnnCONCLUSIONSnBased on structural analysis and mutagenesis data, we propose a previously unappreciated part in beta-arrestins mode of action by which a cationic amphipathic helix may function as a reversible membrane anchor. This novel activation mechanism would facilitate the formation of a high-affinity complex between beta-arrestin and an activated receptor regardless of its specific subtype. Like the interaction between beta-arrestins polar core and the phosphorylated receptor, such a general activation mechanism would contribute to beta-arrestins versatility as a regulator of many receptors.


Journal of Biological Chemistry | 1999

HOW DOES ARRESTIN RESPOND TO THE PHOSPHORYLATED STATE OF RHODOPSIN

Sergey A. Vishnivetskiy; Cherlton L. Paz; Carsten Schubert; Joel A. Hirsch; Paul B. Sigler; Vsevolod V. Gurevich

Visual arrestin quenches light-induced signaling by binding to light-activated, phosphorylated rhodopsin (P-Rh*). Here we present structure-function data, which in conjunction with the refined crystal structure of arrestin (Hirsch, J. A., Schubert, C., Gurevich, V. V., and Sigler, P. B. (1999)Cell, in press), support a model for the conversion of a basal or “inactive” conformation of free arrestin to one that can bind to and inhibit the light activated receptor. The trigger for this transition is an interaction of the phosphorylated COOH-terminal segment of the receptor with arrestin that disrupts intramolecular interactions, including a hydrogen-bonded network of buried, charged side chains, referred to as the “polar core.” This disruption permits structural adjustments that allow arrestin to bind to the receptor. Our mutational survey identifies residues in arrestin (Arg175, Asp30, Asp296, Asp303, Arg382), which when altered bypass the need for the interaction with the receptor’s phosphopeptide, enabling arrestin to bind to activated, nonphosphorylated rhodopsin (Rh*). These mutational changes disrupt interactions and substructures which the crystallographic model and previous biochemical studies have shown are responsible for maintaining the inactive state. The molecular basis for these disruptions was confirmed by successfully introducing structure-based second site substitutions that restored the critical interactions. The nearly absolute conservation of the mutagenically sensitive residues throughout the arrestin family suggests that this mechanism is likely to be applicable to arrestin-mediated desensitization of most G-protein-coupled receptors.


Neuron | 2005

Light-dependent redistribution of arrestin in vertebrate rods is an energy-independent process governed by protein-protein interactions

K. Saidas Nair; Susan M. Hanson; Ana Mendez; Eugenia V. Gurevich; Matthew J. Kennedy; Valery I. Shestopalov; Sergey A. Vishnivetskiy; Jeannie Chen; James B. Hurley; Vsevolod V. Gurevich; Vladlen Z. Slepak

In rod photoreceptors, arrestin localizes to the outer segment (OS) in the light and to the inner segment (IS) in the dark. Here, we demonstrate that redistribution of arrestin between these compartments can proceed in ATP-depleted photoreceptors. Translocation of transducin from the IS to the OS also does not require energy, but depletion of ATP or GTP inhibits its reverse movement. A sustained presence of activated rhodopsin is required for sequestering arrestin in the OS, and the rate of arrestin relocalization to the OS is determined by the amount and the phosphorylation status of photolyzed rhodopsin. Interaction of arrestin with microtubules is increased in the dark. Mutations that enhance arrestin-microtubule binding attenuate arrestin translocation to the OS. These results indicate that the distribution of arrestin in rods is controlled by its dynamic interactions with rhodopsin in the OS and microtubules in the IS and that its movement occurs by simple diffusion.


Journal of Biological Chemistry | 2011

Monomeric Rhodopsin Is Sufficient for Normal Rhodopsin Kinase (GRK1) Phosphorylation and Arrestin-1 Binding

Timothy H. Bayburt; Sergey A. Vishnivetskiy; Mark A. McLean; Takefumi Morizumi; Chih Chin Huang; John J. G. Tesmer; Oliver P. Ernst; Stephen G. Sligar; Vsevolod V. Gurevich

G-protein-coupled receptor (GPCR) oligomerization has been observed in a wide variety of experimental contexts, but the functional significance of this phenomenon at different stages of the life cycle of class A GPCRs remains to be elucidated. Rhodopsin (Rh), a prototypical class A GPCR of visual transduction, is also capable of forming dimers and higher order oligomers. The recent demonstration that Rh monomer is sufficient to activate its cognate G protein, transducin, prompted us to test whether the same monomeric state is sufficient for rhodopsin phosphorylation and arrestin-1 binding. Here we show that monomeric active rhodopsin is phosphorylated by rhodopsin kinase (GRK1) as efficiently as rhodopsin in the native disc membrane. Monomeric phosphorylated light-activated Rh (P-Rh*) in nanodiscs binds arrestin-1 essentially as well as P-Rh* in native disc membranes. We also measured the affinity of arrestin-1 for P-Rh* in nanodiscs using a fluorescence-based assay and found that arrestin-1 interacts with monomeric P-Rh* with low nanomolar affinity and 1:1 stoichiometry, as previously determined in native disc membranes. Thus, similar to transducin activation, rhodopsin phosphorylation by GRK1 and high affinity arrestin-1 binding only requires a rhodopsin monomer.


Journal of Biological Chemistry | 2000

An Additional Phosphate-binding Element in Arrestin Molecule IMPLICATIONS FOR THE MECHANISM OF ARRESTIN ACTIVATION

Sergey A. Vishnivetskiy; Carsten Schubert; Gregory C. Climaco; Yulia V. Gurevich; Maria-Gabriela Velez; Vsevolod V. Gurevich

Arrestins quench the signaling of a wide variety of G protein-coupled receptors by virtue of high-affinity binding to phosphorylated activated receptors. The high selectivity of arrestins for this particular functional form of receptor ensures their timely binding and dissociation. In a continuing effort to elucidate the molecular mechanisms responsible for arrestins selectivity, we used the visual arrestin model to probe the functions of its N-terminal β-strand I comprising the highly conserved hydrophobic element Val-Ile-Phe (residues 11–13) and the adjacent positively charged Lys14 and Lys15. Charge elimination and reversal in positions 14 and 15 dramatically reduce arrestin binding to phosphorylated light-activated rhodopsin (P-Rh*). The same mutations in the context of various constitutively active arrestin mutants (which bind to P-Rh*, dark phosphorylated rhodopsin (P-Rh), and unphosphorylated light-activated rhodopsin (Rh*)) have minimum impact on P-Rh* and Rh* binding and virtually eliminate P-Rh binding. These results suggest that the two lysines “guide” receptor-attached phosphates toward the phosphorylation-sensitive trigger Arg175 and participate in phosphate binding in the active state of arrestin. The elimination of the hydrophobic side chains of residues 11–13 (triple mutation V11A, I12A, and F13A) moderately enhances arrestin binding to P-Rh and Rh*. The effects of triple mutation V11A, I12A, and F13A in the context of phosphorylation-independent mutants suggest that residues 11–13 play a dual role. They stabilize arrestins basal conformation via interaction with hydrophobic elements in arrestins C-tail and α-helix I as well as its active state by interactions with alternative partners. In the context of the recently solved crystal structure of arrestins basal state, these findings allow us to propose a model of initial phosphate-driven structural rearrangements in arrestin that ultimately result in its transition into the active receptor-binding state.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Each rhodopsin molecule binds its own arrestin

Susan Hanson; Eugenia V. Gurevich; Sergey A. Vishnivetskiy; Mohamed R. Ahmed; Xiufeng Song; Vsevolod V. Gurevich

Arrestins (Arrs) are ubiquitous regulators of the most numerous family of signaling proteins, G protein-coupled receptors. Two models of the Arr–receptor interaction have been proposed: the binding of one Arr to an individual receptor or to two receptors in a dimer. To determine the binding stoichiometry in vivo, we used rod photoreceptors where rhodopsin (Rh) and Arr are expressed at comparably high levels and where Arr localization in the light is determined by its binding to activated Rh. Genetic manipulation of the expression of both proteins shows that the maximum amount of Arr that moves to the Rh-containing compartment exceeds 80%, but not 100%, of the molar amount of Rh present. In vitro experiments with purified proteins confirm that Arr “saturates” Rh at a 1:1 ratio. Thus, a single Rh molecule is necessary and sufficient to bind Arr. Remarkable structural conservation among receptors and Arrs strongly suggests that all Arr subtypes bind individual molecules of their cognate receptors.


Journal of Biological Chemistry | 2007

Regulation of Arrestin Binding by Rhodopsin Phosphorylation Level

Sergey A. Vishnivetskiy; Dayanidhi Raman; J. Wei; Matthew J. Kennedy; James B. Hurley; Vsevolod V. Gurevich

Arrestins ensure the timely termination of receptor signaling. The role of rhodopsin phosphorylation in visual arrestin binding was established more than 20 years ago, but the effects of the number of receptor-attached phosphates on this interaction remain controversial. Here we use purified rhodopsin fractions with carefully quantified content of individual phosphorylated rhodopsin species to elucidate the impact of phosphorylation level on arrestin interaction with three biologically relevant functional forms of rhodopsin: light-activated and dark phosphorhodopsin and phospho-opsin. We found that a single receptor-attached phosphate does not facilitate arrestin binding, two are necessary to induce high affinity interaction, and three phosphates fully activate arrestin. Higher phosphorylation levels do not increase the stability of arrestin complex with light-activated rhodopsin but enhance its binding to the dark phosphorhodopsin and phospho-opsin. The complex of arrestin with hyperphosphorylated light-activated rhodopsin is less sensitive to high salt and appears to release retinal faster. These data suggest that arrestin likely quenches rhodopsin signaling after the third phosphate is added by rhodopsin kinase. The complex of arrestin with heavily phosphorylated rhodopsin, which appears to form in certain disease states, has distinct characteristics that may contribute to the phenotype of these visual disorders.


Journal of Biological Chemistry | 2006

Visual and Both Non-visual Arrestins in Their “Inactive” Conformation Bind JNK3 and Mdm2 and Relocalize Them from the Nucleus to the Cytoplasm

Xiufeng Song; Dayanidhi Raman; Eugenia V. Gurevich; Sergey A. Vishnivetskiy; Vsevolod V. Gurevich

Arrestins bind active phosphorylated G protein-coupled receptors, terminating G protein activation. Receptor-bound non-visual arrestins interact with numerous partners, redirecting signaling to alternative pathways. Arrestins also have nuclear localization and nuclear exclusion signals and shuttle between the nucleus and the cytoplasm. Constitutively shuttling proteins often redistribute their interaction partners between the two compartments. Here we took advantage of the nucleoplasmic shuttling of free arrestins and used a “nuclear exclusion assay” to study their interactions with two proteins involved in “life-and-death” decisions in the cell, the kinase JNK3 and the ubiquitin ligase Mdm2. In human embryonic kidney 293 cells green fluorescent protein (GFP)-JNK3 and GFP-Mdm2 predominantly localize in the nucleus, whereas visual arrestin, arrestin2(Q394L) mutant equipped with the nuclear exclusion signal, and arrestin3 localize exclusively to the cytoplasm. Coexpression of arrestins moves both GFP-JNK3 and GFP-Mdm2 to the cytoplasm. Arrestin mutants “frozen” in the basal conformation are the most efficacious. Thus, arrestins in their basal state interact with JNK3 and Mdm2, suggesting that arrestins are likely “preloaded” with their interaction partners when they bind the receptor. Robust interaction of free arrestins with JNK3 and Mdm2 and their ability to regulate subcellular localization of these proteins may play an important role in the survival of photoreceptors and other neurons, as well as in retinal and neuronal degeneration.


The EMBO Journal | 2007

Structure and function of the visual arrestin oligomer

Susan M. Hanson; Ned Van Eps; Derek J. Francis; Christian Altenbach; Sergey A. Vishnivetskiy; Vadim Y. Arshavsky; Candice S. Klug; Wayne L. Hubbell; Vsevolod V. Gurevich

A distinguishing feature of rod arrestin is its ability to form oligomers at physiological concentrations. Using visible light scattering, we show that rod arrestin forms tetramers in a cooperative manner in solution. To investigate the structure of the tetramer, a nitroxide side chain (R1) was introduced at 18 different positions. The effects of R1 on oligomer formation, EPR spectra, and inter‐spin distance measurements all show that the structures of the solution and crystal tetramers are different. Inter‐subunit distance measurements revealed that only arrestin monomer binds to light‐activated phosphorhodopsin, whereas both monomer and tetramer bind microtubules, which may serve as a default arrestin partner in dark‐adapted photoreceptors. Thus, the tetramer likely serves as a ‘storage’ form of arrestin, increasing the arrestin‐binding capacity of microtubules while readily dissociating to supply active monomer when it is needed to quench rhodopsin signaling.

Collaboration


Dive into the Sergey A. Vishnivetskiy'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

Candice S. Klug

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Derek J. Francis

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Miyeon Kim

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge