Network


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

Hotspot


Dive into the research topics where Valentina Tereshko is active.

Publication


Featured researches published by Valentina Tereshko.


Nature | 2008

Structural basis for the function and inhibition of an influenza virus proton channel

Amanda L. Stouffer; Rudresh Acharya; David Salom; Anna S. Levine; Luigi Di Costanzo; Cinque S. Soto; Valentina Tereshko; Vikas Nanda; Steven Stayrook; William F. DeGrado

The M2 protein from influenza A virus is a pH-activated proton channel that mediates acidification of the interior of viral particles entrapped in endosomes. M2 is the target of the anti-influenza drugs amantadine and rimantadine; recently, resistance to these drugs in humans, birds and pigs has reached more than 90% (ref. 1). Here we describe the crystal structure of the transmembrane-spanning region of the homotetrameric protein in the presence and absence of the channel-blocking drug amantadine. pH-dependent structural changes occur near a set of conserved His and Trp residues that are involved in proton gating. The drug-binding site is lined by residues that are mutated in amantadine-resistant viruses. Binding of amantadine physically occludes the pore, and might also perturb the pKa of the critical His residue. The structure provides a starting point for solving the problem of resistance to M2-channel blockers.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Nanoliter microfluidic hybrid method for simultaneous screening and optimization validated with crystallization of membrane proteins

Liang Li; Debarshi Mustafi; Qiang Fu; Valentina Tereshko; Delai L. Chen; Joshua D. Tice; Rustem F. Ismagilov

High-throughput screening and optimization experiments are critical to a number of fields, including chemistry and structural and molecular biology. The separation of these two steps may introduce false negatives and a time delay between initial screening and subsequent optimization. Although a hybrid method combining both steps may address these problems, miniaturization is required to minimize sample consumption. This article reports a “hybrid” droplet-based microfluidic approach that combines the steps of screening and optimization into one simple experiment and uses nanoliter-sized plugs to minimize sample consumption. Many distinct reagents were sequentially introduced as ≈140-nl plugs into a microfluidic device and combined with a substrate and a diluting buffer. Tests were conducted in ≈10-nl plugs containing different concentrations of a reagent. Methods were developed to form plugs of controlled concentrations, index concentrations, and incubate thousands of plugs inexpensively and without evaporation. To validate the hybrid method and demonstrate its applicability to challenging problems, crystallization of model membrane proteins and handling of solutions of detergents and viscous precipitants were demonstrated. By using 10 μl of protein solution, ≈1,300 crystallization trials were set up within 20 min by one researcher. This method was compatible with growth, manipulation, and extraction of high-quality crystals of membrane proteins, demonstrated by obtaining high-resolution diffraction images and solving a crystal structure. This robust method requires inexpensive equipment and supplies, should be especially suitable for use in individual laboratories, and could find applications in a number of areas that require chemical, biochemical, and biological screening and optimization.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Crystal structure of full-length KcsA in its closed conformation

Serdar Uysal; Valeria Vásquez; Valentina Tereshko; Kaori Esaki; Frederic A. Fellouse; Sachdev S. Sidhu; Shohei Koide; Eduardo Perozo; Anthony A. Kossiakoff

KcsA is a proton-activated, voltage-modulated K+ channel that has served as the archetype pore domain in the Kv channel superfamily. Here, we have used synthetic antigen-binding fragments (Fabs) as crystallographic chaperones to determine the structure of full-length KcsA at 3.8 Å, as well as that of its isolated C-terminal domain at 2.6 Å. The structure of the full-length KcsA–Fab complex reveals a well-defined, 4-helix bundle that projects ≈70 Å toward the cytoplasm. This bundle promotes a ≈15° bending in the inner bundle gate, tightening its diameter and shifting the narrowest point 2 turns of helix below. Functional analysis of the full-length KcsA–Fab complex suggests that the C-terminal bundle remains whole during gating. We suggest that this structure likely represents the physiologically relevant closed conformation of KcsA.


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

High-affinity single-domain binding proteins with a binary-code interface

Akiko Koide; Ryan N. Gilbreth; Kaori Esaki; Valentina Tereshko; Shohei Koide

High degrees of sequence and conformation complexity found in natural protein interaction interfaces are generally considered essential for achieving tight and specific interactions. However, it has been demonstrated that specific antibodies can be built by using an interface with a binary code consisting of only Tyr and Ser. This surprising result might be attributed to yet undefined properties of the antibody scaffold that uniquely enhance its capacity for target binding. In this work we tested the generality of the binary-code interface by engineering binding proteins based on a single-domain scaffold. We show that Tyr/Ser binary-code interfaces consisting of only 15–20 positions within a fibronectin type III domain (FN3; 95 residues) are capable of producing specific binding proteins (termed “monobodies”) with a low-nanomolar Kd. A 2.35-Å x-ray crystal structure of a monobody in complex with its target, maltose-binding protein, and mutation analysis revealed dominant contributions of Tyr residues to binding as well as striking molecular mimicry of a maltose-binding protein substrate, β-cyclodextrin, by the Tyr/Ser binary interface. This work suggests that an interaction interface with low chemical diversity but with significant conformational diversity is generally sufficient for tight and specific molecular recognition, providing fundamental insights into factors governing protein–protein interactions.


Journal of the American Chemical Society | 2008

X-ray structure of snow flea antifreeze protein determined by racemic crystallization of synthetic protein enantiomers

Brad L. Pentelute; Zachary P. Gates; Valentina Tereshko; Jennifer L. Dashnau; Jane M. Vanderkooi; Anthony A. Kossiakoff; Stephen B. H. Kent

Chemical protein synthesis and racemic protein crystallization were used to determine the X-ray structure of the snow flea antifreeze protein (sfAFP). Crystal formation from a racemic solution containing equal amounts of the chemically synthesized proteins d-sfAFP and l-sfAFP occurred much more readily than for l-sfAFP alone. More facile crystal formation also occurred from a quasi-racemic mixture of d-sfAFP and l-Se-sfAFP, a chemical protein analogue that contains an additional -SeCH2- moiety at one residue and thus differs slightly from the true enantiomer. Multiple wavelength anomalous dispersion (MAD) phasing from quasi-racemate crystals was then used to determine the X-ray structure of the sfAFP protein molecule. The resulting model was used to solve by molecular replacement the X-ray structure of l-sfAFP to a resolution of 0.98 A. The l-sfAFP molecule is made up of six antiparallel left-handed PPII helixes, stacked in two sets of three, to form a compact brick-like structure with one hydrophilic face and one hydrophobic face. This is a novel experimental protein structure and closely resembles a structural model proposed for sfAFP. These results illustrate the utility of total chemical synthesis combined with racemic crystallization and X-ray crystallography for determining the unknown structure of a protein.


Biopolymers | 1998

X-ray crystallographic analysis of the hydration of A- and B-form DNA at atomic resolution.

Martin Egli; Valentina Tereshko; Marianna Teplova; George Minasov; Andrzei Joachimiak; Ruslan Sanishvili; Charles M. Weeks; Russ Miller; Martin Maier; Haoyun An; P. Dan Cook; Muthiah Manoharan

We have determined single crystal structures of an A-DNA decamer and a B-DNA dodecamer at 0.83 and 0.95 A, respectively. The resolution of the former is the highest reported thus far for any right-handed nucleic acid duplex and the quality of the diffraction data allowed determination of the structure with direct methods. The structures reveal unprecedented details of DNA fine structure and hydration; in particular, we have reexamined the overall hydration of A- and B-form DNA, the distribution of water around phosphate groups, and features of the water structure that may underlie the B to A transition.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Synthetic antibodies for specific recognition and crystallization of structured RNA

Jing-Dong Ye; Valentina Tereshko; John K. Frederiksen; Akiko Koide; Frederic A. Fellouse; Sachdev S. Sidhu; Shohei Koide; Anthony A. Kossiakoff; Joseph A. Piccirilli

Antibodies that bind protein antigens are indispensable in biochemical research and modern medicine. However, knowledge of RNA-binding antibodies and their application in the ever-growing RNA field is lacking. Here we have developed a robust approach using a synthetic phage-display library to select specific antigen-binding fragments (Fabs) targeting a large functional RNA. We have solved the crystal structure of the first Fab–RNA complex at 1.95 Å. Capability in phasing and crystal contact formation suggests that the Fab provides a potentially valuable crystal chaperone for RNA. The crystal structure reveals that the Fab achieves specific RNA binding on a shallow surface with complementarity-determining region (CDR) sequence diversity, length variability, and main-chain conformational plasticity. The Fab–RNA interface also differs significantly from Fab–protein interfaces in amino acid composition and light-chain participation. These findings yield valuable insights for engineering of Fabs as RNA-binding modules and facilitate further development of Fabs as possible therapeutic drugs and biochemical tools to explore RNA biology.


Nature Structural & Molecular Biology | 2001

Crystal structures of the catalytic domain of human protein kinase associated with apoptosis and tumor suppression.

Valentina Tereshko; Marianna Teplova; Joseph S. Brunzelle; D. Martin Watterson; Martin Egli

We have determined X-ray crystal structures with up to 1.5 Å resolution of the catalytic domain of death-associated protein kinase (DAPK), the first described member of a novel family of pro-apoptotic and tumor-suppressive serine/threonine kinases. The geometry of the active site was studied in the apo form, in a complex with nonhydrolyzable AMPPnP and in a ternary complex consisting of kinase, AMPPnP and either Mg2+ or Mn2+. The structures revealed a previously undescribed water-mediated stabilization of the interaction between the lysine that is conserved in protein kinases and the β- and γ-phosphates of ATP, as well as conformational changes at the active site upon ion binding. Comparison between these structures and nucleotide triphosphate complexes of several other kinases disclosed a number of unique features of the DAPK catalytic domain, among which is a highly ordered basic loop in the N-terminal domain that may participate in enzyme regulation.


Protein Science | 2008

Toward chaperone-assisted crystallography: protein engineering enhancement of crystal packing and X-ray phasing capabilities of a camelid single-domain antibody (VHH) scaffold

Valentina Tereshko; Serdar Uysal; Akiko Koide; Katrina Margalef; Shohei Koide; Anthony A. Kossiakoff

A crystallization chaperone is an auxiliary protein that binds to a target of interest, enhances and modulates crystal packing, and provides high‐quality phasing information. We critically evaluated the effectiveness of a camelid single‐domain antibody (VHH) as a crystallization chaperone. By using a yeast surface display system for VHH, we successfully introduced additional Met residues in the core of the VHH scaffold. We identified a set of SeMet‐labeled VHH variants that collectively produced six new crystal forms as the complex with the model antigen, RNase A. The crystals exhibited monoclinic, orthorhombic, triclinic, and tetragonal symmetry and have one or two complexes in the asymmetric unit, some of which diffracted to an atomic resolution. The phasing power of the Met‐enriched VHH chaperone allowed for auto‐building the entire complex using single‐anomalous dispersion technique (SAD) without the need for introducing SeMet into the target protein. We show that phases produced by combining SAD and VHH model‐based phases are accurate enough to easily solve structures of the size reported here, eliminating the need to collect multiple wavelength multiple‐anomalous dispersion (MAD) data. Together with the presence of high‐throughput selection systems (e.g., phage display libraries) for VHH, the enhanced VHH domain described here will be an excellent scaffold for producing effective crystallization chaperones.


Protein Science | 2009

Racemic crystallography of synthetic protein enantiomers used to determine the X-ray structure of plectasin by direct methods

Kalyaneswar Mandal; Brad L. Pentelute; Valentina Tereshko; Vilasak Thammavongsa; Olaf Schneewind; Anthony A. Kossiakoff; Stephen B. H. Kent

We describe the use of racemic crystallography to determine the X‐ray structure of the natural product plectasin, a potent antimicrobial protein recently isolated from fungus. The protein enantiomers L‐plectasin and D‐plectasin were prepared by total chemical synthesis; interestingly, L‐plectasin showed the expected antimicrobial activity, while D‐plectasin was devoid of such activity. The mirror image proteins were then used for racemic crystallization. Synchrotron X‐ray diffraction data were collected to atomic resolution from a racemic plectasin crystal; the racemate crystallized in the achiral centrosymmetric space group P1 with one L‐plectasin molecule and one D‐plectasin molecule forming the unit cell. Dimer‐like intermolecular interactions between the protein enantiomers were observed, which may account for the observed extremely low solvent content (13%–15%) and more highly ordered nature of the racemic crystals. The structure of the plectasin molecule was well defined for all 40 amino acids and was generally similar to the previously determined NMR structure, suggesting minimal impact of the crystal packing on the plectasin conformation.

Collaboration


Dive into the Valentina Tereshko's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marianna Teplova

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrzej Joachimiak

Argonne National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Rustem F. Ismagilov

California Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge