Network


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

Hotspot


Dive into the research topics where Shane Atwell is active.

Publication


Featured researches published by Shane Atwell.


Journal of Biological Chemistry | 2004

A novel mode of Gleevec binding is revealed by the structure of spleen tyrosine kinase

Shane Atwell; Jason M. Adams; John Badger; Michelle D. Buchanan; Ingeborg Feil; Karen Froning; Xia Gao; Jorg Hendle; Kevin Keegan; Barbara Chie Leon; Hans J. Müller-Dieckmann; Vicki Nienaber; Brian Noland; Kai W. Post; K. R. Rajashankar; Aurora Ramos; Marijane Russell; Stephen K. Burley; Sean Buchanan

Spleen tyrosine kinase (Syk) is a non-receptor tyrosine kinase required for signaling from immunoreceptors in various hematopoietic cells. Phosphorylation of two tyrosine residues in the activation loop of the Syk kinase catalytic domain is necessary for signaling, a phenomenon typical of tyrosine kinase family members. Syk in vitro enzyme activity, however, does not depend on phosphorylation (activation loop tyrosine → phenylalanine mutants retain catalytic activity). We have determined the x-ray structure of the unphosphorylated form of the kinase catalytic domain of Syk. The enzyme adopts a conformation of the activation loop typically seen only in activated, phosphorylated tyrosine kinases, explaining why Syk does not require phosphorylation for activation. We also demonstrate that Gleevec (STI-571, Imatinib) inhibits the isolated kinase domains of both unphosphorylated Syk and phosphorylated Abl with comparable potency. Gleevec binds Syk in a novel, compact cis-conformation that differs dramatically from the binding mode observed with unphosphorylated Abl, the more Gleevec-sensitive form of Abl. This finding suggests the existence of two distinct Gleevec binding modes: an extended, trans-conformation characteristic of tight binding to the inactive conformation of a protein kinase and a second compact, cis-conformation characteristic of weaker binding to the active conformation. Finally, the Syk-bound cis-conformation of Gleevec bears a striking resemblance to the rigid structure of the nonspecific, natural product kinase inhibitor staurosporine.


Nature Biotechnology | 2014

Generation of bispecific IgG antibodies by structure-based design of an orthogonal Fab interface

Steven M. Lewis; Xiufeng Wu; Anna Pustilnik; Arlene Sereno; Flora Huang; Heather L Rick; Gurkan Guntas; Andrew Leaver-Fay; Eric Michael Smith; Carolyn Ho; Christophe Hansen-Estruch; Aaron Keith Chamberlain; Stephanie Marie Eaton Truhlar; Elaine M. Conner; Shane Atwell; Brian Kuhlman; Stephen J. Demarest

Robust generation of IgG bispecific antibodies has been a long-standing challenge. Existing methods require extensive engineering of each individual antibody, discovery of common light chains, or complex and laborious biochemical processing. Here we combine computational and rational design approaches with experimental structural validation to generate antibody heavy and light chains with orthogonal Fab interfaces. Parental monoclonal antibodies incorporating these interfaces, when simultaneously co-expressed, assemble into bispecific IgG with improved heavy chain–light chain pairing. Bispecific IgGs generated with this approach exhibit pharmacokinetic and other desirable properties of native IgG, but bind target antigens monovalently. As such, these bispecific reagents may be useful in many biotechnological applications.


Molecular Cancer Therapeutics | 2009

SGX523 is an exquisitely selective, ATP-competitive inhibitor of the MET receptor tyrosine kinase with antitumor activity in vivo

Sean Buchanan; Jorg Hendle; P.S Lee; C.R Smith; P.Y Bounaud; Katayoun Jessen; Crystal Tang; N.H Huser; Jeremy Felce; Karen Froning; M.C Peterman; Brandon E. Aubol; Steven Gessert; J.M Sauder; Kenneth D. Schwinn; Marijane Russell; Isabelle Rooney; Joseph A. Adams; Barbara Chie Leon; T.H Do; Jeff Blaney; P.A Sprengeler; Devon A. Thompson; L Smyth; L.A Pelletier; Shane Atwell; Kevin Holme; Stephen R. Wasserman; Spencer Emtage; Stephen K. Burley

The MET receptor tyrosine kinase has emerged as an important target for the development of novel cancer therapeutics. Activation of MET by mutation or gene amplification has been linked to kidney, gastric, and lung cancers. In other cancers, such as glioblastoma, autocrine activation of MET has been demonstrated. Several classes of ATP-competitive inhibitor have been described, which inhibit MET but also other kinases. Here, we describe SGX523, a novel, ATP-competitive kinase inhibitor remarkable for its exquisite selectivity for MET. SGX523 potently inhibited MET with an IC50 of 4 nmol/L and is >1,000-fold selective versus the >200-fold selectivity of other protein kinases tested in biochemical assays. Crystallographic study revealed that SGX523 stabilizes MET in a unique inactive conformation that is inaccessible to other protein kinases, suggesting an explanation for the selectivity. SGX523 inhibited MET-mediated signaling, cell proliferation, and cell migration at nanomolar concentrations but had no effect on signaling dependent on other protein kinases, including the closely related RON, even at micromolar concentrations. SGX523 inhibition of MET in vivo was associated with the dose-dependent inhibition of growth of tumor xenografts derived from human glioblastoma and lung and gastric cancers, confirming the dependence of these tumors on MET catalytic activity. Our results show that SGX523 is the most selective inhibitor of MET catalytic activity described to date and is thus a useful tool to investigate the role of MET kinase in cancer without the confounding effects of promiscuous protein kinase inhibition. [Mol Cancer Ther 2009;8(12):3181–90]


Protein Science | 2010

Thermal unfolding studies show the disease causing F508del mutation in CFTR thermodynamically destabilizes nucleotide-binding domain 1

Irina I. Protasevich; Zhengrong Yang; Chi Wang; Shane Atwell; Xun Zhao; Spencer Emtage; Diana R. Wetmore; John F. Hunt; Christie G. Brouillette

Misfolding and degradation of CFTR is the cause of disease in patients with the most prevalent CFTR mutation, an in‐frame deletion of phenylalanine (F508del), located in the first nucleotide‐binding domain of human CFTR (hNBD1). Studies of (F508del)CFTR cellular folding suggest that both intra‐ and inter‐domain folding is impaired. (F508del)CFTR is a temperature‐sensitive mutant, that is, lowering growth temperature, improves both export, and plasma membrane residence times. Yet, paradoxically, F508del does not alter the fold of isolated hNBD1 nor did it seem to perturb its unfolding transition in previous isothermal chemical denaturation studies. We therefore studied the in vitro thermal unfolding of matched hNBD1 constructs ±F508del to shed light on the defective folding mechanism and the basis for the thermal instability of (F508del)CFTR. Using primarily differential scanning calorimetry (DSC) and circular dichroism, we show for all hNBD1 pairs studied, that F508del lowers the unfolding transition temperature (Tm) by 6–7°C and that unfolding occurs via a kinetically‐controlled, irreversible transition in isolated monomers. A thermal unfolding mechanism is derived from nonlinear least squares fitting of comprehensive DSC data sets. All data are consistent with a simple three‐state thermal unfolding mechanism for hNBD1 ± F508del: N(±MgATP) ⇄ IT(±MgATP) → AT → (AT)n. The equilibrium unfolding to intermediate, IT, is followed by the rate‐determining, irreversible formation of a partially folded, aggregation‐prone, monomeric state, AT, for which aggregation to (AT)n and further unfolding occur with no detectable heat change. Fitted parameters indicate that F508del thermodynamically destabilizes the native state, N, and accelerates the formation of AT.


Protein Engineering Design & Selection | 2010

Structures of a minimal human CFTR first nucleotide- binding domain as a monomer, head-to-tail homodimer, and pathogenic mutant

Shane Atwell; Christie G. Brouillette; Kris Conners; Spencer Emtage; Tarun Gheyi; William B. Guggino; Jorg Hendle; John F. Hunt; Hal A. Lewis; Frances Lu; Irina I. Protasevich; Logan Rodgers; Rich Romero; Stephen R. Wasserman; Patricia C. Weber; Diana R. Wetmore; Feiyu F. Zhang; Xun Zhao

Upon removal of the regulatory insert (RI), the first nucleotide binding domain (NBD1) of human cystic fibrosis transmembrane conductance regulator (CFTR) can be heterologously expressed and purified in a form that remains stable without solubilizing mutations, stabilizing agents or the regulatory extension (RE). This protein, NBD1 387-646(Delta405-436), crystallizes as a homodimer with a head-to-tail association equivalent to the active conformation observed for NBDs from symmetric ATP transporters. The 1.7-A resolution X-ray structure shows how ATP occupies the signature LSGGQ half-site in CFTR NBD1. The DeltaF508 version of this protein also crystallizes as a homodimer and differs from the wild-type structure only in the vicinity of the disease-causing F508 deletion. A slightly longer construct crystallizes as a monomer. Comparisons of the homodimer structure with this and previously published monomeric structures show that the main effect of ATP binding at the signature site is to order the residues immediately preceding the signature sequence, residues 542-547, in a conformation compatible with nucleotide binding. These residues likely interact with a transmembrane domain intracellular loop in the full-length CFTR channel. The experiments described here show that removing the RI from NBD1 converts it into a well-behaved protein amenable to biophysical studies yielding deeper insights into CFTR function.


Protein Science | 2010

Integrated biophysical studies implicate partial unfolding of NBD1 of CFTR in the molecular pathogenesis of F508del cystic fibrosis

Chi Wang; Irina I. Protasevich; Zhengrong Yang; Derek Seehausen; Timothy Skalak; Xun Zhao; Shane Atwell; J. Spencer Emtage; Diana R. Wetmore; Christie G. Brouillette; John F. Hunt

The lethal genetic disease cystic fibrosis is caused predominantly by in‐frame deletion of phenylalanine 508 in the cystic fibrosis transmembrane conductance regulator (CFTR). F508 is located in the first nucleotide‐binding domain (NBD1) of CFTR, which functions as an ATP‐gated chloride channel on the cell surface. The F508del mutation blocks CFTR export to the surface due to aberrant retention in the endoplasmic reticulum. While it was assumed that F508del interferes with NBD1 folding, biophysical studies of purified NBD1 have given conflicting results concerning the mutations influence on domain folding and stability. We have conducted isothermal (this paper) and thermal (accompanying paper) denaturation studies of human NBD1 using a variety of biophysical techniques, including simultaneous circular dichroism, intrinsic fluorescence, and static light‐scattering measurements. These studies show that, in the absence of ATP, NBD1 unfolds via two sequential conformational transitions. The first, which is strongly influenced by F508del, involves partial unfolding and leads to aggregation accompanied by an increase in tryptophan fluorescence. The second, which is not significantly influenced by F508del, involves full unfolding of NBD1. Mg‐ATP binding delays the first transition, thereby offsetting the effect of F508del on domain stability. Evidence suggests that the initial partial unfolding transition is partially responsible for the poor in vitro solubility of human NBD1. Second‐site mutations that increase the solubility of isolated F508del‐NBD1 in vitro and suppress the trafficking defect of intact F508del‐CFTR in vivo also stabilize the protein against this transition, supporting the hypothesize that it is responsible for the pathological trafficking of F508del‐CFTR.


Methods of Molecular Biology | 2008

High Throughput Protein Production and Crystallization at NYSGXRC

Michael Sauder; Marc E. Rutter; Kevin Bain; Isabelle Rooney; Tarun Gheyi; Shane Atwell; Devon A. Thompson; Spencer Emtage; Stephen K. Burley

Phase II of the Protein Structure Initiative, funded by the NIH NIGMS (National Institute of General Medical Sciences), is a 5-year effort to determine thousands of protein structures. The New York SGX Research Center for Structural Genomics (NYSGXRC) is one of the four large-scale production centers tasked with determining 100-200 structures annually. Almost all protein production is carried out using the high throughput structural biology platform at SGX Pharmaceuticals (SGX), which supplies 120 or more ultrapure proteins per month for NYSGXRC crystallization and structure determination activities. Protocols for PCR, cloning, expression/solubility testing, fermentation, purification, and crystallization are described. General protocols and detailed experimental results for each target are updated weekly at the public PepcDB website (pepcdb.pdb.org/), and all NYSGXRC clones should be available in 2008 through the PlasmID resource operated by the Harvard Institute of Proteomics.


Journal of Medicinal Chemistry | 2015

Synthesis and Pharmacological Characterization of C4-Disubstituted Analogs of 1S,2S,5R,6S-2-Aminobicyclo[3.1.0]hexane-2,6-dicarboxylate: Identification of a Potent, Selective Metabotropic Glutamate Receptor Agonist and Determination of Agonist-Bound Human mGlu2 and mGlu3 Amino Terminal Domain Structures.

James A. Monn; Lourdes Prieto; Lorena Taboada; Concepcion Pedregal; Junliang Hao; Matt R. Reinhard; Steven S. Henry; Paul J. Goldsmith; Christopher David Beadle; Lesley Walton; Teresa Man; Helene Rudyk; Barry Peter Clark; David Edward Tupper; S. Richard Baker; Carlos Lamas; Carlos Montero; Alicia Marcos; Jaime Blanco; Mark G. Bures; David K. Clawson; Shane Atwell; Frances Lu; Jing Wang; Marijane Russell; Beverly A. Heinz; Xushan Wang; Joan H. Carter; Chuanxi Xiang; John T. Catlow

As part of our ongoing research to identify novel agents acting at metabotropic glutamate 2 (mGlu2) and 3 (mGlu3) receptors, we have previously reported the identification of the C4α-methyl analog of mGlu2/3 receptor agonist 1 (LY354740). This molecule, 1S,2S,4R,5R,6S-2-amino-4-methylbicyclo[3.1.0]hexane-2,6-dicarboxylate 2 (LY541850), exhibited an unexpected mGlu2 agonist/mGlu3 antagonist pharmacological profile, whereas the C4β-methyl diastereomer (3) possessed dual mGlu2/3 receptor agonist activity. We have now further explored this structure-activity relationship through the preparation of cyclic and acyclic C4-disubstituted analogs of 1, leading to the identification of C4-spirocyclopropane 5 (LY2934747), a novel, potent, and systemically bioavailable mGlu2/3 receptor agonist which exhibits both antipsychotic and analgesic properties in vivo. In addition, through the combined use of protein-ligand X-ray crystallography employing recombinant human mGlu2/3 receptor amino terminal domains, molecular modeling, and site-directed mutagenesis, a molecular basis for the observed pharmacological profile of compound 2 is proposed.


Structure | 2016

Computationally Designed Bispecific Antibodies using Negative State Repertoires.

Andrew Leaver-Fay; Karen J. Froning; Shane Atwell; Hector Aldaz; Anna Pustilnik; Frances Lu; Flora Huang; Richard Yuan; Saleema Hassanali; Aaron Keith Chamberlain; Jonathan R. Fitchett; Stephen J. Demarest; Brian Kuhlman

A challenge in the structure-based design of specificity is modeling the negative states, i.e., the complexes that you do not want to form. This is a difficult problem because mutations predicted to destabilize the negative state might be accommodated by small conformational rearrangements. To overcome this challenge, we employ an iterative strategy that cycles between sequence design and protein docking in order to build up an ensemble of alternative negative state conformations for use in specificity prediction. We have applied our technique to the design of heterodimeric CH3 interfaces in the Fc region of antibodies. Combining computationally and rationally designed mutations produced unique designs with heterodimer purities greater than 90%. Asymmetric Fc crystallization was able to resolve the interface mutations; the heterodimer structures confirmed that the interfaces formed as designed. With these CH3 mutations, and those made at the heavy-/light-chain interface, we demonstrate one-step synthesis of four fully IgG-bispecific antibodies.


Journal of Medicinal Chemistry | 2015

Synthesis and Pharmacological Characterization of C4-(Thiotriazolyl)-substituted-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylates. Identification of (1R,2S,4R,5R,6R)-2-Amino-4-(1H-1,2,4-triazol-3-ylsulfanyl)bicyclo[3.1.0]hexane-2,6-dicarboxylic Acid (LY2812223), a Highly Potent, Functionally Selective mGlu2 Receptor Agonist.

James A. Monn; Lourdes Prieto; Lorena Taboada; Junliang Hao; Matthew Robert Reinhard; Steven S. Henry; Christopher David Beadle; Lesley Walton; Teresa Man; Helene Rudyk; Barry Peter Clark; David Edward Tupper; S. Richard Baker; Carlos Lamas; Carlos Montero; Alicia Marcos; Jaime Blanco; Mark G. Bures; David K. Clawson; Shane Atwell; Frances Lu; Jing Wang; Marijane Russell; Beverly A. Heinz; Xushan Wang; Joan H. Carter; Brian G. Getman; John T. Catlow; Steven Swanson; Bryan G. Johnson

Identification of orthosteric mGlu(2/3) receptor agonists capable of discriminating between individual mGlu2 and mGlu3 subtypes has been highly challenging owing to the glutamate-site sequence homology between these proteins. Herein we detail the preparation and characterization of a series of molecules related to (1S,2S,5R,6S)-2-aminobicyclo[3.1.0]hexane-2,6-dicarboxylate 1 (LY354740) bearing C4-thiotriazole substituents. On the basis of second messenger responses in cells expressing other recombinant human mGlu2/3 subtypes, a number of high potency and efficacy mGlu2 receptor agonists exhibiting low potency mGlu3 partial agonist/antagonist activity were identified. From this, (1R,2S,4R,5R,6R)-2-amino-4-(1H-1,2,4-triazol-3-ylsulfanyl)bicyclo[3.1.0]hexane-2,6-dicarboxylic acid 14a (LY2812223) was further characterized. Cocrystallization of 14a with the amino terminal domains of hmGlu2 and hmGlu3 combined with site-directed mutation studies has clarified the underlying molecular basis of this unique pharmacology. Evaluation of 14a in a rat model responsive to mGlu2 receptor activation coupled with a measure of central drug disposition provides evidence that this molecule engages and activates central mGlu2 receptors in vivo.

Collaboration


Dive into the Shane Atwell's collaboration.

Top Co-Authors

Avatar

Stephen K. Burley

Case Western Reserve University

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

Jing Wang

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge