Network


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

Hotspot


Dive into the research topics where Stacie L. Bulfer is active.

Publication


Featured researches published by Stacie L. Bulfer.


Science | 2016

2.3 A Resolution Cryo-Em Structure of Human P97 and Mechanism of Allosteric Inhibition

Soojay Banerjee; Alberto Bartesaghi; Alan Merk; Prashant Rao; Stacie L. Bulfer; Yongzhao Yan; Neal Green; Barbara Mroczkowski; R. Jeffrey Neitz; Peter Wipf; Veronica Falconieri; Raymond J. Deshaies; Jacqueline L. S. Milne; Donna M. Huryn; Michelle R. Arkin; Sriram Subramaniam

AAA ATPase conformational high jinks The protein p97 is an AAA adenosine triphosphatase (ATPase) that uses energy from ATP hydrolysis to regulate substrates involved in intracellular protein quality control. Its role in this central process makes it a target for cancer chemotherapy. Banerjee et al. used cryo-electron microscopy to determine high-resolution structures for multiple conformational states of this dynamic macromolecular machine. They also determined the structure of the ADP-bound state bound to an inhibitor. The structures give insight into nucleotide-driven structural changes that drive function and show how inhibitor binding prevents these conformational changes Science, this issue p. 871 Cryo–electron microscopy reveals atomic-resolution structures of a protein complex that is a target for cancer drug development. p97 is a hexameric AAA+ adenosine triphosphatase (ATPase) that is an attractive target for cancer drug development. We report cryo–electron microscopy (cryo-EM) structures for adenosine diphosphate (ADP)–bound, full-length, hexameric wild-type p97 in the presence and absence of an allosteric inhibitor at resolutions of 2.3 and 2.4 angstroms, respectively. We also report cryo-EM structures (at resolutions of ~3.3, 3.2, and 3.3 angstroms, respectively) for three distinct, coexisting functional states of p97 with occupancies of zero, one, or two molecules of adenosine 5′-O-(3-thiotriphosphate) (ATPγS) per protomer. A large corkscrew-like change in molecular architecture, coupled with upward displacement of the N-terminal domain, is observed only when ATPγS is bound to both the D1 and D2 domains of the protomer. These cryo-EM structures establish the sequence of nucleotide-driven structural changes in p97 at atomic resolution. They also enable elucidation of the binding mode of an allosteric small-molecule inhibitor to p97 and illustrate how inhibitor binding at the interface between the D1 and D2 domains prevents propagation of the conformational changes necessary for p97 function.


Journal of Molecular Biology | 2014

Specific inhibition of p97/VCP ATPase and kinetic analysis demonstrate interaction between D1 and D2 ATPase domains.

Tsui Fen Chou; Stacie L. Bulfer; Conrad C. Weihl; Kelin Li; Lev Lis; Michael A. Walters; Frank J. Schoenen; Henry J. Lin; Raymond J. Deshaies; Michelle R. Arkin

The p97 AAA (ATPase associated with diverse cellular activities), also called VCP (valosin-containing protein), is an important therapeutic target for cancer and neurodegenerative diseases. p97 forms a hexamer composed of two AAA domains (D1 and D2) that form two stacked rings and an N-terminal domain that binds numerous cofactor proteins. The interplay between the three domains in p97 is complex, and a deeper biochemical understanding is needed in order to design selective p97 inhibitors as therapeutic agents. It is clear that the D2 ATPase domain hydrolyzes ATP in vitro, but whether D1 contributes to ATPase activity is controversial. Here, we use Walker A and B mutants to demonstrate that D1 is capable of hydrolyzing ATP and show for the first time that nucleotide binding in the D2 domain increases the catalytic efficiency (kcat/Km) of D1 ATP hydrolysis 280-fold, by increasing kcat 7-fold and decreasing Km about 40-fold. We further show that an ND1 construct lacking D2 but including the linker between D1 and D2 is catalytically active, resolving a conflict in the literature. Applying enzymatic observations to small-molecule inhibitors, we show that four p97 inhibitors (DBeQ, ML240, ML241, and NMS-873) have differential responses to Walker A and B mutations, to disease-causing IBMPFD mutations, and to the presence of the N domain binding cofactor protein p47. These differential effects provide the first evidence that p97 cofactors and disease mutations can alter p97 inhibitor potency and suggest the possibility of developing context-dependent inhibitors of p97.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Altered cofactor regulation with disease-associated p97/VCP mutations

Xiaoyi Zhang; Lin Gui; Xiaoyan Zhang; Stacie L. Bulfer; Valentina Sanghez; Daniel E. Wong; You Jin Lee; Lynn W. Lehmann; James Siho Lee; Pei Yin Shih; Henry J. Lin; Michelina Iacovino; Conrad C. Weihl; Michelle R. Arkin; Yanzhuang Wang; Tsui Fen Chou

Significance Age-associated degenerative diseases have similar pathogenic mechanisms related to defects in protein homeostasis. p97/VCP (valosin-containing protein) is essential for coordinating protein degradation and is mutated in a multisystem degenerative disease that affects the central nervous system, muscle, and bone. p97/VCP is an enzyme in the AAA ATPases (ATPases associated with diverse cellular activities) family, which takes apart ATP and uses this energy to perform pivotal functions. We found that p97/VCP cofactors control its enzymatic activity. p97/VCP disease mutants behave abnormally due to lack of appropriate control by these cofactors. Correcting the function of the disease-associated proteins may be a desirable approach to developing safe treatment for fatal degenerative diseases. The next steps are to screen and characterize large panels of compounds to identify potential drugs that may correct the malfunction. Dominant mutations in p97/VCP (valosin-containing protein) cause a rare multisystem degenerative disease with varied phenotypes that include inclusion body myopathy, Paget’s disease of bone, frontotemporal dementia, and amyotrophic lateral sclerosis. p97 disease mutants have altered N-domain conformations, elevated ATPase activity, and altered cofactor association. We have now discovered a previously unidentified disease-relevant functional property of p97 by identifying how the cofactors p37 and p47 regulate p97 ATPase activity. We define p37 as, to our knowledge, the first known p97-activating cofactor, which enhances the catalytic efficiency (kcat/Km) of p97 by 11-fold. Whereas both p37 and p47 decrease the Km of ATP in p97, p37 increases the kcat of p97. In contrast, regulation by p47 is biphasic, with decreased kcat at low levels but increased kcat at higher levels. By deleting a region of p47 that lacks homology to p37 (amino acids 69–92), we changed p47 from an inhibitory cofactor to an activating cofactor, similar to p37. Our data suggest that cofactors regulate p97 ATPase activity by binding to the N domain. Induced conformation changes affect ADP/ATP binding at the D1 domain, which in turn controls ATPase cycling. Most importantly, we found that the D2 domain of disease mutants failed to be activated by p37 or p47. Our results show that cofactors play a critical role in controlling p97 ATPase activity, and suggest that lack of cofactor-regulated communication may contribute to p97-associated disease pathogenesis.


ACS Medicinal Chemistry Letters | 2015

Structure-Activity Study of Bioisosteric Trifluoromethyl and Pentafluorosulfanyl Indole Inhibitors of the AAA ATPase p97.

Celeste Alverez; Michelle R. Arkin; Stacie L. Bulfer; Raffaele Colombo; Marina Kovaliov; Matthew G. LaPorte; Chaemin Lim; Mary Liang; William Jay Moore; R. Jeffrey Neitz; Yongzhao Yan; Zhizhou Yue; Donna M. Huryn; Peter Wipf

Exploratory SAR studies of a new phenyl indole chemotype for p97 inhibition revealed C-5 indole substituent effects in the ADPGlo assay that did not fully correlate with either electronic or steric factors. A focused series of methoxy-, trifluoromethoxy-, methyl-, trifluoromethyl-, pentafluorosulfanyl-, and nitro-analogues was found to exhibit IC50s from low nanomolar to double-digit micromolar. Surprisingly, we found that the trifluoromethoxy-analogue was biochemically a better match of the trifluoromethyl-substituted lead structure than a pentafluorosulfanyl-analogue. Moreover, in spite of their almost equivalent strongly electron-depleting effect on the indole core, pentafluorosulfanyl- and nitro-derivatives were found to exhibit a 430-fold difference in p97 inhibitory activities. Conversely, the electronically divergent C-5 methyl- and nitro-analogues both showed low nanomolar activities.


ACS Medicinal Chemistry Letters | 2016

Allosteric Indole Amide Inhibitors of p97: Identification of a Novel Probe of the Ubiquitin Pathway

Celeste Alverez; Stacie L. Bulfer; Ramappa Chakrasali; Michael S. Chimenti; Raymond J. Deshaies; Neal Green; Mark J. S. Kelly; Matthew G. LaPorte; Taber S. Lewis; Mary Liang; William Jay Moore; R. Jeffrey Neitz; Vsevolod A. Peshkov; Michael A. Walters; Feng Zhang; Michelle R. Arkin; Peter Wipf; Donna M. Huryn

A high-throughput screen to discover inhibitors of p97 ATPase activity identified an indole amide that bound to an allosteric site of the protein. Medicinal chemistry optimization led to improvements in potency and solubility. Indole amide 3 represents a novel uncompetitive inhibitor with excellent physical and pharmaceutical properties that can be used as a starting point for drug discovery efforts.


ACS Chemical Biology | 2016

p97 Disease Mutations Modulate Nucleotide-Induced Conformation to Alter Protein–Protein Interactions

Stacie L. Bulfer; Tsui-Fen Chou; Michelle R. Arkin

The AAA+ ATPase p97/VCP adopts at least three conformations that depend on the binding of ADP and ATP and alter the orientation of the N-terminal protein–protein interaction (PPI) domain into “up” and “down” conformations. Point mutations that cause multisystem proteinopathy 1 (MSP1) are found at the interface of the N domain and D1-ATPase domain and potentially alter the conformational preferences of p97. Additionally, binding of “adaptor” proteins to the N-domain regulates p97’s catalytic activity. We propose that p97/adaptor PPIs are coupled to p97 conformational states. We evaluated the binding of nucleotides and the adaptor proteins p37 and p47 to wild-type p97 and MSP1 mutants. Notably, p47 and p37 bind 8-fold more weakly to the ADP-bound conformation of wild-type p97 compared to the ATP-bound conformation. However, MSP1 mutants lose this nucleotide-induced conformational coupling because they destabilize the ADP-bound, “down” conformation of the N-domain. Loss in conformation coupling to PPIs could contribute to the mechanism of MSP1.


Journal of Biomolecular Screening | 2015

A Fragment-Based Ligand Screen Against Part of a Large Protein Machine The ND1 Domains of the AAA+ ATPase p97/VCP

Michael S. Chimenti; Stacie L. Bulfer; R. Jeffrey Neitz; Adam R. Renslo; Matthew P. Jacobson; Thomas L. James; Michelle R. Arkin; Mark J. S. Kelly

The ubiquitous AAA+ ATPase p97 functions as a dynamic molecular machine driving several cellular processes. It is essential in regulating protein homeostasis, and it represents a potential drug target for cancer, particularly when there is a greater reliance on the endoplasmic reticulum–associated protein degradation pathway and ubiquitin–proteasome pathway to degrade an overabundance of secreted proteins. Here, we report a case study for using fragment-based ligand design approaches against this large and dynamic hexamer, which has multiple potential binding sites for small molecules. A screen of a fragment library was conducted by surface plasmon resonance (SPR) and followed up by nuclear magnetic resonance (NMR), two complementary biophysical techniques. Virtual screening was also carried out to examine possible binding sites for the experimental hits and evaluate the potential utility of fragment docking for this target. Out of this effort, 13 fragments were discovered that showed reversible binding with affinities between 140 µM and 1 mM, binding stoichiometries of 1:1 or 2:1, and good ligand efficiencies. Structural data for fragment–protein interactions were obtained with residue-specific [U-2H] 13CH3-methyl-labeling NMR strategies, and these data were compared to poses from docking. The combination of virtual screening, SPR, and NMR enabled us to find and validate a number of interesting fragment hits and allowed us to gain an understanding of the structural nature of fragment binding.


ACS Medicinal Chemistry Letters | 2018

Optimization of Phenyl Indole Inhibitors of the AAA ATPase p97

Matthew G. LaPorte; James C. Burnett; Raffaele Colombo; Stacie L. Bulfer; Celeste Alverez; Tsui-Fen Chou; R. Jeffrey Neitz; Neal Green; William Jay Moore; Zhizhou Yue; Shan Li; Michelle R. Arkin; Peter Wipf; Donna M. Huryn

Optimization of the side-chain of a phenyl indole scaffold identified from a high-throughput screening campaign for inhibitors of the AAA+ ATPase p97 is reported. The addition of an N-alkyl piperazine led to high potency of this series in a biochemical assay, activity in cell-based assays, and excellent pharmaceutical properties. Molecular modeling based on a subsequently obtained cryo-EM structure of p97 in complex with a phenyl indole was used to rationalize the potency of these allosteric inhibitors.


Organic and Biomolecular Chemistry | 2017

A threonine turnstile defines a dynamic amphiphilic binding motif in the AAA ATPase p97 allosteric binding site

James C. Burnett; Chaemin Lim; Brian D. Peyser; Lalith P. Samankumara; Marina Kovaliov; Raffaele Colombo; Stacie L. Bulfer; Matthew G. LaPorte; Ann R. Hermone; Connor F. McGrath; Michelle R. Arkin; Rick Gussio; Donna M. Huryn; Peter Wipf


Archive | 2017

Applying Biophysical and Biochemical Methods to the Discovery of Allosteric Modulators of the AAA ATPase p97

Stacie L. Bulfer; Michelle R. Arkin

Collaboration


Dive into the Stacie L. Bulfer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donna M. Huryn

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Peter Wipf

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chaemin Lim

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge