557 results on '"Stuckey, Jeanne"'
Search Results
202. Structure-Based Discoveryof BM-957 as a Potent Small-MoleculeInhibitor of Bcl-2 and Bcl-xL Capable of Achieving CompleteTumor Regression.
- Author
-
Chen, Jianfang, Zhou, Haibin, Aguilar, Angelo, Liu, Liu, Bai, Longchuan, McEachern, Donna, Yang, Chao-Yie, Meagher, Jennifer L., Stuckey, Jeanne A., and Wang, Shaomeng
- Published
- 2012
- Full Text
- View/download PDF
203. Structure-Based Designof Potent Bcl-2/Bcl-xL Inhibitorswith Strong in Vivo Antitumor Activity.
- Author
-
Zhou, Haibin, Aguilar, Angelo, Chen, Jianfang, Bai, Longchuan, Liu, Liu, Meagher, Jennifer L., Yang, Chao-Yie, McEachern, Donna, Cong, Xin, Stuckey, Jeanne A., and Wang, Shaomeng
- Published
- 2012
- Full Text
- View/download PDF
204. Design of Bcl-2 and Bcl-xLInhibitors with SubnanomolarBinding Affinities Based upon a New Scaffold.
- Author
-
Zhou, Haibin, Chen, Jianfang, Meagher, Jennifer L., Yang, Chao-Yie, Aguilar, Angelo, Liu, Liu, Bai, Longchuan, Cong, Xin, Cai, Qian, Fang, Xueliang, Stuckey, Jeanne A., and Wang, Shaomeng
- Published
- 2012
- Full Text
- View/download PDF
205. Structural basis for the enhanced stability of highly fluorinated proteins.
- Author
-
Buer, Benjamin C., Meagher, Jennifer L., Stuckey, Jeanne A., and Marsh, E. Neil G.
- Subjects
PROTEIN structure ,AMINO acids ,FLUORINATION ,PROTEIN stability ,FLUOROCARBONS ,HYDROCARBONS - Abstract
Noncanonical amino acids have proved extremely useful for modifying the properties of proteins. Among them, extensively fluorinated (fluorous) amino acids seem particularly effective in increasing protein stability; however, in the absence of structural data, the basis of this stabilizing effect remains poorly understood. To address this problem, we solved X-ray structures for three small proteins with hydrophobic cores that are packed with either fluorocarbon or hydrocarbon side chains and compared their stabilities. Although larger, the fluorinated residues are accommodated within the protein with minimal structural perturbation, because they closely match the shape of the hydrocarbon side chains that they replace. Thus, stability increases seem to be better explained by increases in buried hydrophobic surface area that accompany fluorination than by specific fluorous interactions between fluorinated side chains. This finding is illustrated by the design of a highly fluorinated protein that, by compensating for the larger volume and surface area of the fluorinated side chains, exhibits similar stability to its nonfluorinated counterpart. These structure-based observations should inform efforts to rationally modulate protein function using noncanonical amino acids. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
206. Hydrolytic catalysis and structural stabilization in a designed metalloprotein.
- Author
-
Zastrow, Melissa L., Peacock, Anna F. A., Stuckey, Jeanne A., and Pecoraro, Vincent L.
- Subjects
METALLOPROTEINS ,CATALYTIC activity ,METALLOENZYMES ,HYDROLYSIS ,METAL ions ,IRON proteins - Abstract
Metal ions are an important part of many natural proteins, providing structural, catalytic and electron transfer functions. Reproducing these functions in a designed protein is the ultimate challenge to our understanding of them. Here, we present an artificial metallohydrolase, which has been shown by X-ray crystallography to contain two different metal ions-a Zn(II) ion, which is important for catalytic activity, and a Hg(II) ion, which provides structural stability. This metallohydrolase displays catalytic activity that compares well with several characteristic reactions of natural enzymes. It catalyses p-nitrophenyl acetate (pNPA) hydrolysis with an efficiency only ~100-fold less than that of human carbonic anhydrase (CA)II and at least 550-fold better than comparable synthetic complexes. Similarly, CO
2 hydration occurs with an efficiency within ~500-fold of CAII. Although histidine residues in the absence of Zn(II) exhibit pNPA hydrolysis, miniscule apopeptide activity is observed for CO2 hydration. The kinetic and structural analysis of this first de novo designed hydrolytic metalloenzyme reveals necessary design features for future metalloenzymes containing one or more metals. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
207. Structural bases for the different anti-fibrillatory effects of chloroquine and quinidine.
- Author
-
Noujaim, Sami F., Stuckey, Jeanne A., Ponce-Balbuena, Daniela, Ferrer-Villada, Tania, López-Izquierdo, Angelica, Pandit, Sandeep V., Sánchez-Chapula, José A., and Jalife, José
- Subjects
- *
MYOCARDIAL depressants , *ANTIMALARIALS , *CHLOROQUINE , *QUINIDINE , *ION channels , *DRUG interactions , *ELECTROPHYSIOLOGY , *PHARMACOKINETICS , *LABORATORY mice - Abstract
Aims Chloroquine, an anti-malarial quinoline, is structurally similar to quinidine. Both drugs have been shown to block ion channels. We tested the hypothesis that chloroquine's mode of interaction with the vestibule of the cytoplasmic domain of the inward rectifier potassium channel Kir2.1 makes it a more effective IK1 blocker and anti-fibrillatory agent than quinidine. Methods and results We used comparative molecular modelling and ligand docking of the three-dimensional structures of quinidine and chloroquine in the intracellular domain of Kir2.1. Simulations predicted that chloroquine effectively blocks potassium flow by binding at the centre of the ion permeation vestibule of Kir2.1. In contrast, quinidine binds the vestibular side, only partially blocking ion movement. We tested the modelling predictions in Kir2.1-expressing human embryonic kidney (HEK)-293 cells. The half-maximal inhibitory concentration for chloroquine block of IK1 was 1.2 µM, while that of quinidine was 57 µM. Finally, we used optical mapping of Langendorff-perfused mouse hearts with cardiac-specific Kir2.1 up-regulation to compare the anti-fibrillatory effects of the drugs. In five of six hearts, 10 μM quinidine slowed the frequency but did not terminate the tachyarrhythmia. In five of five hearts, 10 μM chloroquine terminated the arrhythmia, restoring sinus rhythm. Conclusion Quinidine only partially blocks IK1. Chloroquine binds at the centre of the ion permeation vestibule of Kir2.1, which makes it a more effective IK1 blocker and anti-fibrillatory agent than quinidine. Integrating the structural biology of drug-ion channel interactions with cellular electrophysiology and optical mapping is an excellent approach to understand the molecular mechanisms of anti-arrhythmic drug action and for drug discovery. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
208. Co-operative Versus Independent Transport of Different Cargoes by Kinesin-1.
- Author
-
Hammond, Jennetta W., Griffin, Kelly, Jih, Gloria T., Stuckey, Jeanne, and Verhey, Kristen J.
- Subjects
KINESIN ,MICROTUBULES ,PROTEINS ,BINDING sites ,NEURAL stem cells - Abstract
Kinesin motors drive the intracellular transport of multiple cargoes along microtubule tracks; yet, how kinesins discriminate among their many potential cargoes is unknown. We tested whether Kinesin-1 cargoes compete, co-operate or are transported independently of each other. We focused on Kinesin-1 cargoes that bind directly to the kinesin light chain (KLC) subunit, namely the c-Jun NH
2 -terminal kinase-interacting proteins (JIPs) 1 and 3, Kidins220/ARMS and PAT1. Overexpression of individual cargo proteins in differentiated CAD cells resulted in mislocalization of the endogenous protein but had no effect on localization of other cargo proteins to neurite tips. Thus, while transport of distinct cargoes is saturable, they do not compete with each other. Interestingly, we found that low expression of JIP1 or JIP3 enhanced the transport of the other JIP to neurite tips. Moreover, JIP1 and JIP3 require each other for transport. Co-operative transport is due to an interaction between JIP1 and JIP3 as well as distinct binding sites on the KLC tetratricopeptide repeat (TPR) bundle: the TPR groove binds to C-terminal residues of JIP1, whereas the TPR surface binds to internal residues in JIP3. Formation of a JIP1/JIP3/KLC complex is necessary for efficient JIP1 or JIP3 transport in neuronal cells. Thus, JIP scaffolding proteins are transported in a co-operative manner, despite the independent transport of other Kinesin-1 cargoes. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
209. LplA1-dependent utilization of host lipoyl peptides enables Listeria cytosolic growth and virulence.
- Author
-
Keeney, Kristie M., Stuckey, Jeanne A., and O'Riordan, Mary X. D.
- Subjects
- *
LISTERIA monocytogenes , *PATHOGENIC microorganisms , *CYTOSOL , *LIGASES , *PEPTIDES , *LIPOIC acid , *CELLS - Abstract
The bacterial pathogen Listeria monocytogenes replicates within the cytosol of mammalian cells. Mechanisms by which the bacterium exploits the host cytosolic environment for essential nutrients are poorly defined. L. monocytogenes is a lipoate auxotroph and must scavenge this critical cofactor, using lipoate ligases to facilitate attachment of the lipoyl moiety to metabolic enzyme complexes. Although the L. monocytogenes genome encodes two putative lipoate ligases, LplA1 and LplA2, intracellular replication and virulence require only LplA1. Here we show that LplA1 enables utilization of host-derived lipoyl peptides by L. monocytogenes. LplA1 is dispensable for growth in the presence of free lipoate, but necessary for growth on low concentrations of mammalian lipoyl peptides. Furthermore, we demonstrate that the intracellular growth defect of the Δ lplA1 mutant is rescued by addition of exogenous lipoic acid to host cells, suggesting that L. monocytogenes dependence on LplA1 is dictated by limiting concentrations of available host lipoyl substrates. Thus, the ability of L. monocytogenes and other intracellular pathogens to efficiently use host lipoyl peptides as a source of lipoate may be a requisite adaptation for life within the mammalian cell. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
210. Identifying important structural characteristics of arsenic resistance proteins by using designed three-stranded coiled coils.
- Author
-
Touw, Debra S., Nordman, Christer E., Stuckey, Jeanne A., and Pecoraro, Vincent L.
- Subjects
ARSENIC ,NATIVE element minerals ,WATER pollution ,PROTEINS ,BACTERIA - Abstract
Arsenic, a contaminant of water supplies worldwide, is one of the most toxic inorganic ions. Despite arsenic's health impact, there is relatively little structural detail known about its interactions with proteins. Bacteria such as Escherichia co/i have evolved arsenic resistance using the Ars operon that is regulated by ArsR, a repressor protein that dissociates from DNA when As(III) binds. This protein undergoes a critical conformational change upon binding As(III) with three cysteine residues. Unfortunately, structures of ArsR with or without As(III) have not been reported. Alternatively, de novo designed peptides can bind As(III) in an endo configuration within a thiolate-rich environment consistent with that proposed for both ArsR and ArsD. We report the structure of the As(III) complex of Coil Ser L9C to a 1.8-Å resolution, providing x-ray characterization of As(III) in a Tris thiolate protein environment and allowing a structural basis by which to understand arsenated ArsR. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
211. Laboratory evolution of one disulfide isomerase to resemble another.
- Author
-
Hiniker, Annie, Ren, Guoping, Heras, Begoña, Ying Zheng, Laurinec, Stephanie, Jobson, Richard W., Stuckey, Jeanne A., Martin, Jennifer L., and Bardwell, James C. A.
- Subjects
ISOMERASES ,ASPARTATE aminotransferase ,MOLECULAR chaperones ,PROTEIN folding ,ESCHERICHIA coli ,PROKARYOTES - Abstract
It is often difficult to determine which of the sequence and structural differences between divergent members of multigene families are functionally important. Here we use a laboratory evolution approach to determine functionally important structural differences between two distantly related disulfide isomerases, DsbC and DsbG from Escherichia coil. Surprisingly, we found single amino acid substitutions in DsbG that were able to complement dsbC in vivo and have more DsbC-like isomerase activity in vitro. Crystal structures of the three strongest point mutants, DsbG K113E, DsbG V216M, and DsbG T200M, reveal changes in highly surface-exposed regions that cause DsbG to more closely resemble the distantly related DsbC. In this case, laboratory evolution appears to have taken a direct route to allow one protein family member to complement another, with single substitutions apparently bypassing much of the need for multiple changes that took place over ≈0.5 billion years of evolution. Our findings suggest that, for these two proteins at least, regions important in determining functional differences may represent only a tiny fraction of the overall protein structure. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
212. Two substrate-targeting sites in theYersiniaprotein tyrosine phosphatase co-operate to promote bacterial virulence.
- Author
-
Ivanov, Maya I., Stuckey, Jeanne A., Schubert, Heidi L., Saper, Mark A., and Bliska, James B.
- Subjects
- *
PROTEIN-tyrosine phosphatase , *PHOSPHOPROTEIN phosphatases , *YERSINIA , *ENTEROBACTERIACEAE , *MICROBIAL virulence , *EPITHELIAL cells , *EPITHELIUM , *PATHOGENIC bacteria - Abstract
YopH is a protein tyrosine phosphatase and an essential virulence determinant of the pathogenic bacteriumYersinia. Yersiniadelivers YopH into infected host cells using a type III secretion mechanism. YopH dephosphorylates several focal adhesion proteins including p130Cas in human epithelial cells, resulting in disruption of focal adhesions and cell detachment from the extracellular matrix. How the C-terminal protein tyrosine phosphatase domain of YopH targets specific substrates such as p130Cas in the complex milieu of the host cell has not been fully elucidated. An N-terminal non-catalytic domain of YopH binds p130Cas in a phosphotyrosine-dependent manner and functions as a novel substrate-targeting site. The structure of the YopH protein tyrosine phosphatase domain bound to a model phosphopeptide substrate was solved and the resulting structure revealed a second substrate-targeting site (‘site 2’) within the catalytic domain. Site 2 binds to p130Cas in a phosphotyrosine-dependent manner, and co-operates with the N-terminal domain (‘site 1’) to promote efficient recognition of p130Cas by YopH in epithelial cells. The identification of two substrate-targeting sites in YopH that co-operate to promote epithelial cell detachment and bacterial virulence reinforces the importance of protein–protein interactions for determining protein tyrosine phosphatase specificityin vivo, and highlights the sophisticated nature of microbial pathogenicity factors. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
213. The Role of HTS in Drug Discovery at the University of Michigan
- Author
-
J. Larsen, Martha, D. Larsen, Scott, Fribley, Andrew, Grembecka, Jolanta, Homan, Kristoff, Mapp, Anna, Haak, Andrew, Nikolovska-Coleska, Zaneta, A. Stuckey, Jeanne, Sun, Duxin, and H.Sherman, David
- Abstract
High throughput screening (HTS) is an integral part of a highly collaborative approach to drug discovery at the University of Michigan. The HTS lab is one of four core centers that provide services to identify, produce, screen and follow-up on biomedical targets for faculty. Key features of this system are: protein cloning and purification, protein crystallography, small molecule and siRNA HTS, medicinal chemistry and pharmacokinetics. Therapeutic areas that have been targeted include anti-bacterial, metabolic, neurodegenerative, cardiovascular, anti-cancer and anti-viral. The centers work in a coordinated, interactive environment to affordably provide academic investigators with the technology, informatics and expertise necessary for successful drug discovery. This review provides an overview of these centers at the University of Michigan, along with case examples of successful collaborations with faculty.
- Published
- 2014
214. Crystal structure of a phospholipase D family member.
- Author
-
Stuckey, Jeanne A. and Dixon, Jack E.
- Subjects
- *
CRYSTALS , *PHOSPHOLIPASES - Abstract
The first crystal structure of a phospholipase D (PLD) family member has been determined at 2.0 Å resolution. The PLD superfamily is defined by a common sequence motif, HxK(x)4D(x)6GSxN, and includes enzymes involved in signal transduction, lipid biosynthesis, endonucleases and open reading frames in pathogenic viruses and bacteria. The crystal structure suggests that residues from two sequence motifs form a single active site. A histidine residue from one motif acts as a nucleophile in the catalytic mechanism, forming a phosphoenzyme intermediate, whereas a histidine residue from the other motif appears to function as a general acid in the cleavage of the phosphodiester bond. The structure suggests that the conserved lysine residues are involved in phosphate binding. Large-scale genomic sequencing revealed that there are many PLD family members. Our results suggest that all of these proteins may possess a common structure and catalytic mechanism. [ABSTRACT FROM AUTHOR]
- Published
- 1999
215. Crystal structure of Yersinia protein tyrosine phosphatase at 2.5A and the complex with tungstate.
- Author
-
Stuckey, Jeanne A. and Schubert, Heidi L.
- Subjects
- *
PHOSPHATASES , *YERSINIA , *CHEMICAL structure - Abstract
Presents the structures of the unliganded and tungstate-bound crystal forms of protein tyrosine phosphatases (PTPase). Ribbon diagram of the unliganded Yersinia PTPase structure; Yersinia PTPase P-loop and hydrogen-bonding arrays.
- Published
- 1994
- Full Text
- View/download PDF
216. Form and function in protein dephosphorylation.
- Author
-
Denu, John M. and Stuckey, Jeanne A.
- Subjects
- *
PHOSPHOPROTEIN phosphatases - Abstract
Focuses on the structure and function of the protein phosphatases. Structure of serine/threonine protein phosphatases; Structure of protein tyrosine phosphatases; Molecular reaction mechanism.
- Published
- 1996
- Full Text
- View/download PDF
217. Cloning, Expression, and Characterization of the Galα1,3Gal High Affinity Lectin from the Mushroom Marasmius oreades*
- Author
-
Kruger, Robert P., Winter, Harry C., Simonson-Leff, Nancy, Stuckey, Jeanne A., Goldstein, Irwin J., and Dixon, Jack E.
- Abstract
The purification and unique carbohydrate binding properties, including blood group B-specific agglutination and preferential binding to Galα1,3Gal-containing sugar epitopes, of theMarasmius oreadesagglutinin (MOA) are reported in an accompanying paper (Winter, H. C., Mostafapour, K., and Goldstein, I. J. (2002) J. Biol. Chem.277, 14996–15001). Here we describe the cloning, characterization, and expression of MOA. MOA was digested with trypsin and endoproteinase Asp-N, and the peptide fragments were purified by high performance liquid chromatography. Amino acid sequence data were obtained for eight peptides. Using oligonucleotides deduced from the peptide sequences for a reverse transcriptase-PCR, a 41-base pair cDNA was obtained. The 41-base pair fragment allowed the generation a full-length cDNA using 5′ and 3′ rapid amplification of cDNA ends. MOA cDNA encodes a protein of 293 amino acids that contains a ricin domain. These carbohydrate binding domains were first described in subunits of bacterial toxins and are also commonly found in polysaccharide-degrading enzymes. Whereas these proteins are known to display a variety of sugar binding specificities, none to date are known to share MOA's high affinity for Galα1,3Gal and Galα1,3Galβ1,4GlcNAc. Recombinantly expressed and purified MOA retains the specificity and affinity observed with the native protein. This study provides the basis for analyzing the underlying cause for the unusual binding specificity of MOA.
- Published
- 2002
- Full Text
- View/download PDF
218. A Unique Carbohydrate Binding Domain Targets the Lafora Disease Phosphatase to Glycogen*
- Author
-
Wang, Jianyong, Stuckey, Jeanne A., Wishart, Matthew J., and Dixon, Jack E.
- Abstract
Lafora disease (progressive myoclonus epilepsy of Lafora type) is an autosomal recessive neurodegenerative disorder resulting from defects in the EPM2Agene. EPM2Aencodes a 331-amino acid protein containing a carboxyl-terminal phosphatase catalytic domain. We demonstrate that the EPM2Agene product also contains an amino-terminal carbohydrate binding domain (CBD) and that the CBD is critical for association with glycogen both in vitroand in vivo. The CBD domain localizes the phosphatase to specific subcellular compartments that correspond to the expression pattern of glycogen processing enzyme, glycogen synthase. Mutations in the CBD result in mis-localization of the phosphatase and thereby suggest that the CBD targets laforin to intracellular glycogen particles where it is likely to function. Thus naturally occurring mutations within the CBD of laforin likely result in progressive myoclonus epilepsy due to mis-localization of phosphatase expression.
- Published
- 2002
- Full Text
- View/download PDF
219. Interaction between Pyrin and the Apoptotic Speck Protein (ASC) Modulates ASC-induced Apoptosis*
- Author
-
Richards, Neil, Schaner, Philip, Diaz, Arturo, Stuckey, Jeanne, Shelden, Eric, Wadhwa, Anish, and Gumucio, Deborah L.
- Abstract
Patients with familial Mediterranean fever suffer sporadic inflammatory attacks characterized by fever and intense pain (in joints, abdomen, or chest). Pyrin, the product of theMEFVlocus, is a cytosolic protein whose function is unknown. Using pyrin as a “bait” to probe a yeast two-hybrid library made from neutrophil cDNA, we isolatedapoptotic speck protein containing a caspase recruitment domain (CARD) (ASC), a proapoptotic protein that induces the formation of large cytosolic “specks” in transfected cells. We found that when HeLa cells are transfected with ASC, specks are formed. After co-transfection of cells with ASC plus wild type pyrin, an increase in speck-positive cells is found, and speck-positive cells show increased survival. Immunofluorescence studies show that pyrin co-localizes with ASC in specks. Speck localization requires exon 1 of pyrin, but exon 1 alone of pyrin does not result in an increase in the number of specks. Exon 1 of pyrin and exon 1 of ASC show 42% sequence similarity and resemble death domain-related structures in modeling studies. These findings link pyrin to apoptosis pathways and suggest that the modulation of cell survival may be a component of the pathophysiology of familial Mediterranean fever.
- Published
- 2001
- Full Text
- View/download PDF
220. Computational Cosolvent Mapping Analysis Leads to Identify Salicylic Acid Analogs as Weak Inhibitors of ST2 and IL33 Binding
- Author
-
Yuan, Xinrui, Chinnaswamy, Krishnapriya, Stuckey, Jeanne A., and Yang, Chao-Yie
- Abstract
Cytokine signaling initiated by the binding of the cytokine receptors to cytokines plays important roles in immune regulation and diseases. Structurally, cytokine receptors interact with cytokines via an extensive, rugged interface that represents a challenge in inhibitor development. Our computational analysis has previously indicated that butyric acid, mimicking acidic residues, preferentially binds to sites in ST2 (Stimulation-2) that interact with acidic residues of IL33, the endogenous cytokine for ST2. To investigate if a charged group in small molecules facilitates ligand binding to ST2, we developed a biochemical homogeneous time resolved fluorescence assay to determine the inhibition of ST2/IL33 binding by five molecules containing an aromatic ring and a charged group. Three molecules, including niacin, salicylic acid, and benzamidine, exhibit inhibition activities at millimolar concentrations. We further employed the computational cosolvent mapping analysis to identify a shared mode of interaction between niacin, salicylic acid, and ST2. The mode of interaction was further confirmed by four analogous compounds that exhibited similar or improved activities. Our study provided the evidence of inhibition of ST2 and IL33 binding by salicylic acid and analogs. The results suggest that biological activity of salicylic acid may be partly mediated through modulating extracellular cytokine receptors and cytokine interaction.
- Published
- 2022
- Full Text
- View/download PDF
221. The X-ray Crystal Structures of YersiniaTyrosine Phosphatase with Bound Tungstate and Nitrate
- Author
-
Fauman, Eric B., Yuvaniyama, Chantana, Schubert, Heidi L., Stuckey, Jeanne A., and Saper, Mark A.
- Abstract
X-ray crystal structures of the Yersiniatyrosine phosphatase (PTPase) in complex with tungstate and nitrate have been solved to 2.4-Å resolution. Tetrahedral tungstate, WO2−4, is a competitive inhibitor of the enzyme and is isosteric with the substrate and product of the catalyzed reaction. Planar nitrate, NO−3, is isosteric with the PO3moiety of a phosphotransfer transition state. The crystal structures of the YersiniaPTPase with and without ligands, together with biochemical data, permit modeling of key steps along the reaction pathway. These energy-minimized models are consistent with a general acid-catalyzed, in-line displacement of the phosphate moiety to Cys403on the enzyme, followed by attack by a nucleophilic water molecule to release orthophosphate. This nucleophilic water molecule is identified in the crystal structure of the nitrate complex. The active site structure of the PTPase is compared to alkaline phosphatase, which employs a similar phosphomonoester hydrolysis mechanism. Both enzymes must stabilize charges at the nucleophile, the PO3moiety of the transition state, and the leaving group. Both an associative (bond formation preceding bond cleavage) and a dissociative (bond cleavage preceding bond formation) mechanism were modeled, but a dissociative-like mechanism is favored for steric and chemical reasons. Since nearly all of the 47 invariant or highly conserved residues of the PTPase domain are clustered at the active site, we suggest that the mechanism postulated for the Yersiniaenzyme is applicable to all the PTPases.
- Published
- 1996
- Full Text
- View/download PDF
222. Discovery of M-1121 as an Orally Active Covalent Inhibitor of Menin-MLL Interaction Capable of Achieving Complete and Long-Lasting Tumor Regression
- Author
-
Zhang, Meng, Aguilar, Angelo, Xu, Shilin, Huang, Liyue, Chinnaswamy, Krishnapriya, Sleger, Taryn, Wang, Bo, Gross, Stefan, Nicolay, Brandon N., Ronseaux, Sebastien, Harvey, Kaitlin, Wang, Yu, McEachern, Donna, Kirchhoff, Paul D., Liu, Zhaomin, Stuckey, Jeanne, Tron, Adriana E., Liu, Tao, and Wang, Shaomeng
- Abstract
Targeting the menin-MLL protein–protein interaction is being pursued as a new therapeutic strategy for the treatment of acute leukemia carrying MLL-rearrangements (MLLr leukemia). Herein, we report M-1121, a covalent and orally active inhibitor of the menin-MLL interaction capable of achieving complete and persistent tumor regression. M-1121 establishes covalent interactions with Cysteine 329 located in the MLL binding pocket of menin and potently inhibits growth of acute leukemia cell lines carrying MLL translocations with no activity in cell lines with wild-type MLL. Consistent with the mechanism of action, M-1121 drives dose-dependent down-regulation of HOXA9and MEIS1gene expression in the MLL-rearranged MV4;11 leukemia cell line. M-1121 is orally bioavailable and shows potent antitumor activity in vivowith tumor regressions observed at tolerated doses in the MV4;11 subcutaneous and disseminated models of MLL-rearranged leukemia. Together, our findings support development of an orally active covalent menin inhibitor as a new therapy for MLLr leukemia.
- Published
- 2021
- Full Text
- View/download PDF
223. Open Reading Frame 1 Protein of the Human Long Interspersed Nuclear Element 1 Retrotransposon Binds Multiple Equivalents of Lead
- Author
-
Pinter, Tyler B. J., Ruckthong, Leela, Stuckey, Jeanne A., Deb, Aniruddha, Penner-Hahn, James E., and Pecoraro, Vincent L.
- Abstract
The human long interspersed nuclear element 1 (LINE1) has been implicated in numerous diseases and has been suggested to play a significant role in genetic evolution. Open reading frame 1 protein (ORF1p) is one of the two proteins encoded in this self-replicating mobile genetic element, both of which are essential for retrotransposition. The structure of the three-stranded coiled-coil domain of ORF1p was recently solved and showed the presence of tris-cysteine layers in the interior of the coiled-coil that could function as metal binding sites. Here, we demonstrate that ORF1p binds Pb(II). We designed a model peptide, GRCSL16CL23C, to mimic two of the ORF1p Cys3layers and crystallized the peptide both as the apo-form and in the presence of Pb(II). Structural comparison of the ORF1p with apo-(GRCSL16CL23C)3shows very similar Cys3layers, preorganized for Pb(II) binding. We propose that exposure to heavy metals, such as lead, could influence directly the structural parameters of ORF1p and thus impact the overall LINE1 retrotransposition frequency, directly relating heavy metal exposure to genetic modification.
- Published
- 2021
- Full Text
- View/download PDF
224. Discovery of EEDi-5273 as an Exceptionally Potent and Orally Efficacious EED Inhibitor Capable of Achieving Complete and Persistent Tumor Regression
- Author
-
Rej, Rohan Kalyan, Wang, Changwei, Lu, Jianfeng, Wang, Mi, Petrunak, Elyse, Zawacki, Kaitlin P., McEachern, Donna, Yang, Chao-Yie, Wang, Lu, Li, Ruiting, Chinnaswamy, Krishnapriya, Wen, Bo, Sun, Duxin, Stuckey, Jeanne A., Zhou, Yunlong, Chen, Jianyong, Tang, Guozhi, and Wang, Shaomeng
- Abstract
Embryonic ectoderm development (EED) is a promising therapeutic target for human cancers and other diseases. We report herein the discovery of exceptionally potent and efficacious EED inhibitors. By conformational restriction of a previously reported EED inhibitor, we obtained a potent lead compound. Further optimization of the lead yielded exceptionally potent EED inhibitors. The best compound EEDi-5273 binds to EED with an IC50value of 0.2 nM and inhibits the KARPAS422 cell growth with an IC50value of 1.2 nM. It demonstrates an excellent PK and ADME profile, and its oral administration leads to complete and persistent tumor regression in the KARPAS422 xenograft model with no signs of toxicity. Co-crystal structures of two potent EED inhibitors with EED provide a solid structural basis for their high-affinity binding. EEDi-5273 is a promising EED inhibitor for further advanced preclinical development for the treatment of human cancer and other human diseases.
- Published
- 2021
- Full Text
- View/download PDF
225. Engineering a Therapeutic Lectin by Uncoupling Mitogenicity from Antiviral Activity
- Author
-
Kaplan, Mark H., Swanson, Michael D., Gabius, Hans Joachim, Goldstein, Irwin J., Smee, Donald F., Schols, Dominique, Tarbet, E. Bart, King, Steven, Xue, Yi, Hoffmann, Hans Heinrich, Meagher, Jennifer L., Garcia, J. Victor, Boudreaux, Daniel M., Murphy, Paul V., Rice, Charles M., Oscarson, Stefan, Salmon, Loïc, De La Fuente, Cynthia, Winter, Harry C., Markovitz, David M., Roy, René, Stuckey, Jeanne A., André, Sabine, Chugh, Jeetender, Hurst, Brett L., and Al-Hashimi, Hashim M.
- Subjects
3. Good health - Abstract
A key effector route of the Sugar Code involves lectins that exert crucial regulatory controls by targeting distinct cellular glycans. We demonstrate that a single amino acid substitution in a banana lectin, replacing histidine 84 with a threonine, significantly reduces its mitogenicity while preserving its broad-spectrum antiviral potency. X-ray crystallography, NMR spectroscopy, and glycocluster assays reveal that loss of mitogenicity is strongly correlated with loss of pi-pi stacking between aromatic amino acids H84 and Y83, which removes a wall separating two carbohydrate binding sites, thus diminishing multivalent interactions. On the other hand, monovalent interactions and antiviral activity are preserved by retaining other wild-type conformational features and possibly through unique contacts involving the T84 side chain. Through such fine-tuning, target selection and downstream effects of a lectin can be modulated so as to knock down one activity while preserving another, thus providing tools for therapeutics and for understanding the Sugar Code.
226. Development of Novel Small-Molecule Activators of Pyruvate Kinase Muscle Isozyme 2, PKM2, to Reduce Photoreceptor Apoptosis.
- Author
-
Wubben, Thomas J., Chaudhury, Sraboni, Watch, Brennan T., Stuckey, Jeanne A., Weh, Eric, Fernando, Roshini, Goswami, Moloy, Pawar, Mercy, Rech, Jason C., and Besirli, Cagri G.
- Subjects
- *
PYRUVATE kinase , *APOPTOSIS , *PHOTORECEPTORS , *FUNCTIONAL groups , *VISION disorders , *ANILINE - Abstract
Treatment options are lacking to prevent photoreceptor death and subsequent vision loss. Previously, we demonstrated that reprogramming metabolism via the pharmacologic activation of PKM2 is a novel photoreceptor neuroprotective strategy. However, the features of the tool compound used in those studies, ML-265, preclude its advancement as an intraocular, clinical candidate. This study sought to develop the next generation of small-molecule PKM2 activators, aimed specifically for delivery into the eye. Compounds were developed that replaced the thienopyrrolopyridazinone core of ML-265 and modified the aniline and methyl sulfoxide functional groups. Compound 2 demonstrated that structural changes to the ML-265 scaffold are tolerated from a potency and efficacy standpoint, allow for a similar binding mode to the target, and circumvent apoptosis in models of outer retinal stress. To overcome the low solubility and problematic functional groups of ML-265, compound 2's efficacious and versatile core structure for the incorporation of diverse functional groups was then utilized to develop novel PKM2 activators with improved solubility, lack of structural alerts, and retained potency. No other molecules are in the pharmaceutical pipeline for the metabolic reprogramming of photoreceptors. Thus, this study is the first to cultivate the next generation of novel, structurally diverse, small-molecule PKM2 activators for delivery into the eye. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
227. Correction to Design ofBcl-2 and Bcl-xL Inhibitors with Subnanomolar Binding Affinities Basedupon a New Scaffold.
- Author
-
Zhou, Haibin, Chen, Jianfang, Meagher, Jennifer L., Yang, Chao-Yie, Aguilar, Angelo, Liu, Liu, Bai, Longchuan, Cong, Xin, Cai, Qian, Fang, Xueliang, Stuckey, Jeanne A., and Wang, Shaomeng
- Published
- 2012
- Full Text
- View/download PDF
228. Crystal structure of banana lectin reveals a novel second sugar binding site
- Author
-
Meagher, Jennifer L., Winter, Harry C., Ezell, Porscha, Goldstein, Irwin J., and Stuckey, Jeanne A.
- Abstract
Banana lectin (Banlec) is a dimeric plant lectin from the jacalin-related lectin family. Banlec belongs to a subgroup of this family that binds to glucose/mannose, but is unique in recognizing internal α1,3 linkages as well as β1,3 linkages at the reducing termini. Here we present the crystal structures of Banlec alone and with laminaribiose (LAM) (Glcβ1, 3Glc) and Xyl-β1,3-Man-α-O-Methyl. The structure of Banlec has a β-prism-I fold, similar to other family members, but differs from them in its mode of sugar binding. The reducing unit of the sugar is inserted into the binding site causing the second saccharide unit to be placed in the opposite orientation compared with the other ligand-bound structures of family members. More importantly, our structures reveal the presence of a second sugar binding site that has not been previously reported in the literature. The residues involved in the second site are common to other lectins in this family, potentially signaling a new group of mannose-specific jacalin-related lectins (mJRL) with two sugar binding sites.
- Published
- 2005
- Full Text
- View/download PDF
229. Conserved Residues in the HAMP Domain Define a New Family of Proposed Bipartite Energy Taxis Receptors.
- Author
-
Elliott, Kathryn T., Zhulin, Igor B., Stuckey, Jeanne A., and DiRita, Victor J.
- Subjects
- *
CELLULAR signal transduction - Abstract
An abstract of the article "Conserved Residues in the HAMP Domain Define a New Family of Proposed Bipartite Energy Taxis Receptors," by Kathryn T. Elliott, Igor B. Zhulin, Jeanne A. Stuckey and Victor J. DiRita is presented.
- Published
- 2009
- Full Text
- View/download PDF
230. Function of His185 in Aquifex aeolicus 3-Deoxy-d-manno-octulosonate 8-Phosphate Synthase
- Author
-
Wang, Jian, Duewel, Henry S., Stuckey, Jeanne A., Woodard, Ronald W., and Gatti, Domenico L.
- Subjects
- *
ENZYMES , *LIGANDS (Biochemistry) , *3-Deoxy-D-manno-octulosonate 8-phosphate synthase - Abstract
Aquifex aeolicus 3-deoxy-d-manno-octulosonate 8-phosphate synthase (KDO8PS) catalyzes the condensation of arabinose 5-phosphate (A5P) and phosphoenolpyruvate (PEP) by favoring the activation of a water molecule coordinated to the active-site metal ion. Cys11, His185, Glu222 and Asp233 are the other metal ligands. Wild-type KDO8PS is purified with Zn2+ or Fe2+ in the active site, but maximal activity in vitro is achieved when the endogenous metal is replaced with Cd2+. The H185G enzyme retains 8% of the wild-type activity. ICP mass spectrometry analysis indicates that loss of His185 decreases the enzyme affinity for Fe2+, but not for Zn2+. However, maximal activity is again achieved by substitution of the endogenous metal with Cd2+. We have determined the X-ray structures of the Cd2+ H185G enzyme in its substrate-free form, and in complex with PEP, and PEP plus A5P. These structures show a normal amount of Cd2+ bound, suggesting that coordination by His185 is not essential to retain Cd2+ in the active site. Nonetheless, there are significant changes in the coordination sphere of Cd2+ with respect to the wild-type enzyme, as the carboxylate moiety of PEP binds directly to the metal ion and replaces water and His185 as ligands. These observations indicate that the primary function of His185 in A. aeolicus KDO8PS is to orient PEP in the active site of the enzyme in such a way that a water molecule on the sinister (si) side of PEP can be activated by direct coordination to the metal ion. [Copyright &y& Elsevier]
- Published
- 2002
- Full Text
- View/download PDF
231. Lipid-based vaccine nanoparticles for induction of humoral immune responses against HIV-1 and SARS-CoV-2.
- Author
-
Park, Kyung Soo, Bazzill, Joseph D., Son, Sejin, Nam, Jutaek, Shin, Seung Won, Ochyl, Lukasz J., Stuckey, Jeanne A., Meagher, Jennifer L., Chang, Louise, Song, Jun, Montefiori, David C., LaBranche, Celia C., Smith, Janet L., Xu, Jie, and Moon, James J.
- Subjects
- *
HUMORAL immunity , *SARS-CoV-2 , *HIV , *VIRAL antigens , *CORONAVIRUSES - Abstract
The current health crisis of corona virus disease 2019 (COVID-19) highlights the urgent need for vaccine systems that can generate potent and protective immune responses. Protein vaccines are safe, but conventional approaches for protein-based vaccines often fail to elicit potent and long-lasting immune responses. Nanoparticle vaccines designed to co-deliver protein antigens and adjuvants can promote their delivery to antigen-presenting cells and improve immunogenicity. However, it remains challenging to develop vaccine nanoparticles that can preserve and present conformational epitopes of protein antigens for induction of neutralizing antibody responses. Here, we have designed a new lipid-based nanoparticle vaccine platform (NVP) that presents viral proteins (HIV-1 and SARS-CoV-2 antigens) in a conformational manner for induction of antigen-specific antibody responses. We show that NVP was readily taken up by dendritic cells (DCs) and promoted DC maturation and antigen presentation. NVP loaded with BG505.SOSIP.664 (SOSIP) or SARS-CoV-2 receptor-binding domain (RBD) was readily recognized by neutralizing antibodies, indicating the conformational display of antigens on the surfaces of NVP. Rabbits immunized with SOSIP-NVP elicited strong neutralizing antibody responses against HIV-1. Furthermore, mice immunized with RBD-NVP induced robust and long-lasting antibody responses against RBD from SARS-CoV-2. These results suggest that NVP is a promising platform technology for vaccination against infectious pathogens. A nanoparticle vaccine platform that preserves conformational epitopes of protein antigens elicits robust humoral immune responses against HIV-1 and SARS-CoV-2. Unlabelled Image • Vaccine nanoparticles effectively deliver antigens to dendritic cells in draining lymph nodes. • Vaccine nanoparticles allow conformational display of viral antigens from HIV-1 or SARS-CoV-2. • Vaccine nanoparticles elicit strong neutralizing antibody responses against HIV-1 in rabbits. • Vaccine nanoparticles induce robust and long-lasting antibody responses against SARS-CoV-2 in mice. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
232. Targeted disruption of pi–pi stacking in Malaysian banana lectin reduces mitogenicity while preserving antiviral activity.
- Author
-
Covés-Datson, Evelyn M., King, Steven R., Legendre, Maureen, Swanson, Michael D., Gupta, Auroni, Claes, Sandra, Meagher, Jennifer L., Boonen, Arnaud, Zhang, Lihong, Kalveram, Birte, Raglow, Zoe, Freiberg, Alexander N., Prichard, Mark, Stuckey, Jeanne A., Schols, Dominique, and Markovitz, David M.
- Subjects
- *
LECTINS , *ANTIVIRAL agents , *PROTEIN folding , *AMINO acids , *AROMATIC compounds - Abstract
Lectins, carbohydrate-binding proteins, have been regarded as potential antiviral agents, as some can bind glycans on viral surface glycoproteins and inactivate their functions. However, clinical development of lectins has been stalled by the mitogenicity of many of these proteins, which is the ability to stimulate deleterious proliferation, especially of immune cells. We previously demonstrated that the mitogenic and antiviral activities of a lectin (banana lectin, BanLec) can be separated via a single amino acid mutation, histidine to threonine at position 84 (H84T), within the third Greek key. The resulting lectin, H84T BanLec, is virtually non-mitogenic but retains antiviral activity. Decreased mitogenicity was associated with disruption of pi–pi stacking between two aromatic amino acids. To examine whether we could provide further proof-of-principle of the ability to separate these two distinct lectin functions, we identified another lectin, Malaysian banana lectin (Malay BanLec), with similar structural features as BanLec, including pi–pi stacking, but with only 63% amino acid identity, and showed that it is both mitogenic and potently antiviral. We then engineered an F84T mutation expected to disrupt pi–pi stacking, analogous to H84T. As predicted, F84T Malay BanLec (F84T) was less mitogenic than wild type. However, F84T maintained strong antiviral activity and inhibited replication of HIV, Ebola, and other viruses. The F84T mutation disrupted pi–pi stacking without disrupting the overall lectin structure. These findings show that pi–pi stacking in the third Greek key is a conserved mitogenic motif in these two jacalin-related lectins BanLec and Malay BanLec, and further highlight the potential to rationally engineer antiviral lectins for therapeutic purposes. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
233. Structure of the zinc-finger antiviral protein in complex with RNA reveals a mechanism for selective targeting of CG-rich viral sequences.
- Author
-
Meagher, Jennifer L., Takata, Matthew, Gonçalves-Carneiro, Daniel, Keane, Sarah C., Rebendenne, Antoine, Ong, Heley, Orr, Victoria K., MacDonald, Margaret R., Stuckey, Jeanne A., Bieniasz, Paul D., and Smith, Janet L.
- Subjects
- *
ZINC-finger proteins , *RNA , *NUCLEIC acids , *MOLECULAR structure - Abstract
Infection of animal cells by numerous viruses is detected and countered by a variety of means, including recognition of nonself nucleic acids. The zinc finger antiviral protein (ZAP) depletes cytoplasmic RNA that is recognized as foreign in mammalian cells by virtue of its elevated CG dinucleotide content compared with endogenous mRNAs. Here, we determined a crystal structure of a protein-RNA complex containing the N-terminal, 4-zinc finger human (h) ZAP RNA-binding domain (RBD) and a CG dinucleotide-containing RNA target. The structure reveals in molecular detail how hZAP is able to bind selectively to CG-rich RNA. Specifically, the 4 zinc fingers create a basic patch on the hZAP RBD surface. The highly basic second zinc finger contains a pocket that selectively accommodates CG dinucleotide bases. Structure guided mutagenesis, cross-linking immunoprecipitation sequencing assays, and RNA affinity assays show that the structurally defined CG-binding pocket is not required for RNA binding per se in human cells. However, the pocket is a crucial determinant of high-affinity, specific binding to CG dinucleotide-containing RNA. Moreover, variations in RNA-binding specificity among a panel of CG-binding pocket mutants quantitatively predict their selective antiviral activity against a CGenriched HIV-1 strain. Overall, the hZAP RBD RNA structure provides an atomic-level explanation for how ZAP selectively targets foreign, CG-rich RNA. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
234. Crucial Role of the SH2B1 PH Domain for the Control of Energy Balance.
- Author
-
Flores, Anabel, Argetsinger, Lawrence S., Stadler, Lukas K. J., Malaga, Alvaro E., Vander, Paul B., DeSantis, Lauren C., Joe, Ray M., Cline, Joel M., Keogh, Julia M., Henning, Elana, Barroso, Ines, Mendes de Oliveira, Edson, Chandrashekar, Gowri, Clutter, Erik S., Yixin Hu, Stuckey, Jeanne, Farooqi, I. Sadaf, Myers Jr., Martin G., Carter-Su, Christin, and Hu, Yixin
- Subjects
- *
GLUCOSE intolerance , *INSULIN resistance , *ADAPTOR proteins , *HUMAN abnormalities , *OBESITY - Abstract
Disruption of the adaptor protein SH2B1 (SH2-B, PSM) is associated with severe obesity, insulin resistance, and neurobehavioral abnormalities in mice and humans. Here, we identify 15 SH2B1 variants in severely obese children. Four obesity-associated human SH2B1 variants lie in the Pleckstrin homology (PH) domain, suggesting that the PH domain is essential for SH2B1's function. We generated a mouse model of a human variant in this domain (P322S). P322S/P322S mice exhibited substantial prenatal lethality. Examination of the P322S/+ metabolic phenotype revealed late-onset glucose intolerance. To circumvent P322S/P322S lethality, mice containing a two-amino acid deletion within the SH2B1 PH domain (ΔP317, R318 [ΔPR]) were studied. Mice homozygous for ΔPR were born at the expected Mendelian ratio and exhibited obesity plus insulin resistance and glucose intolerance beyond that attributable to their increased adiposity. These studies demonstrate that the PH domain plays a crucial role in how SH2B1 controls energy balance and glucose homeostasis. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
235. Changing the Apoptosis Pathway through Evolutionary Protein Design.
- Author
-
Shultis, David, Mitra, Pralay, Huang, Xiaoqiang, Johnson, Jarrett, Khattak, Naureen Aslam, Gray, Felicia, Piper, Clint, Czajka, Jeff, Hansen, Logan, Wan, Bingbing, Chinnaswamy, Krishnapriya, Liu, Liu, Wang, Mi, Pan, Jingxi, Stuckey, Jeanne, Cierpicki, Tomasz, Borchers, Christoph H., Wang, Shaomeng, Lei, Ming, and Zhang, Yang
- Subjects
- *
APOPTOSIS , *PROTEIN engineering , *ISOTHERMAL titration calorimetry , *PROTEIN binding , *BIOCHEMISTRY experiments - Abstract
Abstract One obstacle in de novo protein design is the vast sequence space that needs to be searched through to obtain functional proteins. We developed a new method using structural profiles created from evolutionarily related proteins to constrain the simulation search process, with functions specified by atomic-level ligand–protein binding interactions. The approach was applied to redesigning the BIR3 domain of the X-linked inhibitor of apoptosis protein (XIAP), whose primary function is to suppress the cell death by inhibiting caspase-9 activity; however, the function of the wild-type XIAP can be eliminated by the binding of Smac peptides. Isothermal calorimetry and luminescence assay reveal that the designed XIAP domains can bind strongly with the Smac peptides but do not significantly inhibit the caspase-9 proteolytic activity in vitro compared with the wild-type XIAP protein. Detailed mutation assay experiments suggest that the binding specificity in the designs is essentially determined by the interplay of structural profile and physical interactions, which demonstrates the potential to modify apoptosis pathways through computational design. Graphical abstract Unlabelled Image Highlights • Potential to modify apoptosis pathways through computational protein design • New protein design algorithm built on evolutionary profiles • Design functional XIAP BIR3 domain using evolutionary profiles • Design binding specificity by combining profile and physical interactions • Testify protein design by hybrid structure prediction and biochemistry experiments [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
236. Structure of cytochrome P450 2B4 with an acetate ligand and an active site hydrogen bond network similar to oxyferrous P450cam.
- Author
-
Yang, Yuting, Bu, Weishu, Im, Sangchoul, Meagher, Jennifer, Stuckey, Jeanne, and Waskell, Lucy
- Subjects
- *
ACETATES , *HYDROGEN bonding , *CYTOCHROME P-450 , *BIOLOGICAL pigments , *MOLECULAR association - Published
- 2018
- Full Text
- View/download PDF
237. Probing the interaction between the histone methyltransferase/deacetylase subunit RBBP4/7 and the transcription factor BCL11A in epigenetic complexes.
- Author
-
Moody, Rebecca Reed, Miao-Chia Lo, Meagher, Jennifer L., Chang-Ching Lin, Stevers, Nicholas O., Tinsley, Samantha L., Inkyung Jung, Matvekas, Aleksas, Stuckey, Jeanne A., and Duxin Sun
- Subjects
- *
TRANSCRIPTION factors , *TRIPLE-negative breast cancer , *METHYLTRANSFERASES , *DEACETYLASES , *HEMATOPOIETIC system - Abstract
The transcription factor BCL11A has recently been reported to be a driving force in triple-negative breast cancer (TNBC), contributing to the maintenance of a chemoresistant breast cancer stem cell (BCSC) population. Although BCL11A was shown to suppress γ-globin and p21 and to induce MDM2 expression in the hematopoietic system, its downstream targets in TNBC are still unclear. For its role in transcriptional repression, BCL11A was found to interact with several corepressor complexes; however, the mechanisms underlying these interactions remain unknown. Here, we reveal that BCL11A interacts with histone methyltransferase (PRC2) and histone deacetylase (NuRD and SIN3A) complexes through their common subunit, RBBP4/7. In fluorescence polarization assays, we show that BCL11A competes with histone H3 for binding to the negatively charged top face of RBBP4. To define that interaction, we solved the crystal structure of RBBP4 in complex with an N-terminal peptide of BCL11A (residues 2-16, BCL11A(2-16)). The crystal structure identifies novel interactions between BCL11A and the side of the β-propeller of RBBP4 that are not seen with histone H3. We next show that BCL11A(2-16) pulls down RBBP4, RBBP7, and other components of PRC2, NuRD, and SIN3A from the cell lysate of theTNBCcell line SUM149. Furthermore, we demonstrate the therapeutic potential of targeting the RBBP4-BCL11A binding by showing that a BCL11A peptide can decrease aldehyde dehydrogenase-positive BCSCs and mammosphere formation capacity in SUM149. Together, our findings have uncovered a previously unidentified mechanism that BCL11A may use to recruit epigenetic complexes to regulate transcription and promote tumorigenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
238. High-resolution crystal structures of Colocasia esculenta tarin lectin.
- Author
-
Pereira, Patricia R., Meagher, Jennifer L., Winter, Harry C., Goldstein, Irwin J., Paschoalin, Vânia M. F., Silva, Joab T., and Stuckey, Jeanne A.
- Subjects
- *
CRYSTAL structure , *TARO , *LECTINS , *HETERODIMERS , *MONOMERS , *AGGLUTININS - Abstract
Tarin, the Colocasia esculenta lectin from the superfamily of α-D-mannose-specific plant bulb lectins, is a tetramer of 47 kDa composed of two heterodimers. Each heterodimer possesses homologous monomers of ~11.9 (A chain) and ~12.7 (B chain) kDa. The structures of apo and carbohydrate-bound tarin were solved to 1.7 Å and 1.91 Å, respectively. Each tarin monomer forms a canonical β-prism II fold, common to all members of Galanthus nivalis agglutinin (GNA) family, which is partially stabilized by a disulfide bond and a conserved hydrophobic core. The heterodimer is formed through domain swapping involving the C-terminal β-strand and the β-sheet on face I of the prism. The tetramer is assembled through the dimerization of the B chains from heterodimers involving face II of each prism. The 1.91 Å crystal structure of tarin bound to Manα(1,3)Manα(1,6)Man reveals an expanded carbohydrate-binding sequence (QxDxNxVxYx4/6WX) on face III of the β-prism. Both monomers possess a similar fold, except for the length of the loop, which begins after the conserved tyrosine and creates the binding pocket for the α(1,6)-terminal mannose. This loop differs in size and amino-acid composition from 10 other β-prism II domain proteins, and may confer carbohydrate-binding specificity among members of the GNA-related lectin family. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
239. D3R grand challenge 2015: Evaluation of protein-ligand pose and affinity predictions.
- Author
-
Gathiaka, Symon, Liu, Shuai, Chiu, Michael, Yang, Huanwang, Stuckey, Jeanne, Kang, You, Delproposto, Jim, Kubish, Ginger, Dunbar, James, Carlson, Heather, Burley, Stephen, Walters, W., Amaro, Rommie, Feher, Victoria, and Gilson, Michael
- Subjects
- *
MOLECULAR docking , *FREE energy (Thermodynamics) , *LIGANDS (Biochemistry) , *PROTEIN receptors , *DRUG design - Abstract
The Drug Design Data Resource (D3R) ran Grand Challenge 2015 between September 2015 and February 2016. Two targets served as the framework to test community docking and scoring methods: (1) HSP90, donated by AbbVie and the Community Structure Activity Resource (CSAR), and (2) MAP4K4, donated by Genentech. The challenges for both target datasets were conducted in two stages, with the first stage testing pose predictions and the capacity to rank compounds by affinity with minimal structural data; and the second stage testing methods for ranking compounds with knowledge of at least a subset of the ligand-protein poses. An additional sub-challenge provided small groups of chemically similar HSP90 compounds amenable to alchemical calculations of relative binding free energy. Unlike previous blinded Challenges, we did not provide cognate receptors or receptors prepared with hydrogens and likewise did not require a specified crystal structure to be used for pose or affinity prediction in Stage 1. Given the freedom to select from over 200 crystal structures of HSP90 in the PDB, participants employed workflows that tested not only core docking and scoring technologies, but also methods for addressing water-mediated ligand-protein interactions, binding pocket flexibility, and the optimal selection of protein structures for use in docking calculations. Nearly 40 participating groups submitted over 350 prediction sets for Grand Challenge 2015. This overview describes the datasets and the organization of the challenge components, summarizes the results across all submitted predictions, and considers broad conclusions that may be drawn from this collaborative community endeavor. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
240. The First Crystal Structure of the UP1 Domain of hnRNP A1 Bound to RNA Reveals a New Look for an Old RNA Binding Protein.
- Author
-
Morgan, Christopher E., Meagher, Jennifer L., Levengood, Jeffrey D., Delproposto, James, Rollins, Carrie, Stuckey, Jeanne A., and Tolbert, Blanton S.
- Subjects
- *
NUCLEOPROTEIN structure , *HETEROGENOUS nucleation , *PROTEIN crystallography , *PROTEIN binding , *SINGLE-stranded DNA , *RNA - Abstract
The heterogeneous nuclear ribonucleoprotein (hnRNP) A1 protein is a multifunctional RNA binding protein implicated in a wide range of biological functions. Mechanisms and putative hnRNP A1–RNA interactions have been inferred primarily from the crystal structure of its UP1 domain bound to ssDNA. RNA stem loops represent an important class of known hnRNP A1 targets, yet little is known about the structural basis of hnRNP A1–RNA recognition. Here, we report the first high-resolution structure (1.92 Å) of UP1 bound to a 5′-AGU-3′ trinucleotide that resembles sequence elements of several native hnRNP A1–RNA stem loop targets. UP1 interacts specifically with the AG dinucleotide sequence via a “nucleobase pocket” formed by the β-sheet surface of RRM1 and the inter-RRM linker; RRM2 does not contact the RNA. The inter-RRM linker forms the lid of the nucleobase pocket and we show using structure-guided mutagenesis that the conserved salt-bridge interactions (R75:D155 and R88:D157) on the α-helical side of the RNA binding surface stabilize the linker in a geometry poised to bind RNA. We further investigated the structural basis of UP1 binding HIV i SL3 ESS3 by determining a structural model of the complex scored by small-angle X-ray scattering. UP1 docks on the apical loop of SL3 ESS3 using its RRM1 domain and inter-RRM linker only. The biophysical implications of the structural model were tested by measuring kinetic binding parameters, where mutations introduced within the apical loop reduce binding affinities by slowing down the rate of complex formation. Collectively, the data presented here provide the first insights into hnRNP A1–RNA interactions. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
241. Structure-Activity relationship of 1-(Furan-2ylmethyl)Pyrrolidine-Based Stimulation-2 (ST2) inhibitors for treating graft versus host disease.
- Author
-
Yuan, Xinrui, Jiang, Hua, Fu, Denggang, Robida, Aaron, Rajanayake, Krishani, Yuan, Hebao, Wen, Bo, Sun, Duxin, Watch, Brennan T., Chinnaswamy, Krishnapriya, Stuckey, Jeanne A., Paczesny, Sophie, Rech, Jason C., and Yang, Chao-Yie
- Subjects
- *
GRAFT versus host disease , *STRUCTURE-activity relationships , *HEMATOPOIETIC stem cell transplantation , *REGULATORY T cells - Abstract
[Display omitted] An elevated plasma level of soluble ST2 (sST2) is a risk biomarker for graft-versus-host disease (GVHD) and death in patients receiving hematopoietic cell transplantation (HCT). sST2 functions as a trap for IL-33 and amplifies the pro-inflammatory type 1 and 17 response while suppressing the tolerogenic type 2 and regulatory T cells activation during GVHD development. We previously identified small-molecule ST2 inhibitors particularly iST2-1 that reduces plasma sST2 levels and improved survival in two animal models. Here, we reported the structure–activity relationship of the furanylmethylpyrrolidine-based ST2 inhibitors based on iST2-1. Based on the biochemical AlphaLISA assay, we improved the activity of iST2-1 by 6-fold (∼6 μM in IC 50 values) in the inhibition of ST2/IL-33 and confirmed the activities of the compounds in a cellular reporter assay. To determine the inhibition of the alloreactivity in vitro, we used the mixed lymphocyte reaction assay to demonstrate that our ST2 inhibitors decreased CD4+ and CD8+ T cells proliferation and increased Treg population. The data presented in this work are critical to the development of ST2 inhibitors in future. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
242. Structural analysis and binding properties of isoforms of tarin, the GNA-related lectin from Colocasia esculenta.
- Author
-
Pereira, Patrícia R., Winter, Harry C., Verícimo, Mauricio A., Meagher, Jennifer L., Stuckey, Jeanne A., Goldstein, Irwin J., Paschoalin, Vânia M.F., and Silva, Joab T.
- Subjects
- *
LECTINS , *TARO , *PROTEIN structure , *PROTEIN binding , *COMMON snowdrop , *PROTEIN stability , *CHEMICAL modification of proteins - Abstract
The lectins, a class of proteins that occur widely in animals, plants, fungi, lichens and microorganisms, are known for their ability to specifically bind to carbohydrates. Plant lectins can be classified into 12 families including the Galanthus nivalis agglutinin (GNA)-related lectin superfamily, which is widespread among monocotyledonous plants and binds specifically to mannose, a behavior that confers remarkable anti-tumor, anti-viral and insecticidal properties on these proteins. The present study characterized a mitogenic lectin from this family, called tarin, which was purified from the crude extract from taro ( Colocasia esculenta ). The results showed that tarin is a glycoprotein with 2–3% carbohydrate content, composed of least 10 isoforms with pIs ranging from 5.5 to 9.5. The intact protein is a heterotetramer of 47 kDa composed of two non-identical and non-covalently associated polypeptides, with small subunits of 11.9 kDa and large subunits of 12.6 kDa. The tarin structure is stable and recovers or maintains its functional structure following treatments at different temperatures and pH. Tarin showed a complex carbohydrate specificity, binding with high affinity to high-mannose and complex N-glycans. Many of these ligands can be found in viruses, tumor cells and insects, as well as in hematopoietic progenitor cells. Chemical modifications confirmed that both conserved and non-conserved amino acids participate in this interaction. This study determined the structural and ligand binding characteristics of a GNA-related lectin that can be exploited for several different purposes, particularly as a proliferative therapeutic molecule that is able to enhance the immunological response. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
243. SAR405838: An Optimized Inhibitor of MDM2-p53 Interaction That Induces Complete and Durable Tumor Regression.
- Author
-
Shaomeng Wang, Wei Sun, Yujun Zhao, McEachern, Donna, Meaux, Isabelle, Barrière, Cédric, Stuckey, Jeanne A., Meagher, Jennifer L., Longchuan Bai, Liu Liu, Hoffman-Luca, Cassandra Gianna, Jianfeng Lu, Shangary, Sanjeev, Shanghai Yu, Bernard, Denzil, Aguilar, Angelo, Dos-Santos, Odette, Besret, Laurent, Guerif, Stéphane, and Pannier, Pascal
- Subjects
- *
P53 protein , *PROTEIN-protein interactions , *SPONTANEOUS cancer regression , *LEUKEMIA treatment , *XENOGRAFTS , *APOPTOSIS - Abstract
Blocking the oncoprotein murine double minute 2 (MDM2)-p53 protein-protein interaction has long been considered to offer a broad cancer therapeutic strategy, despite the potential risks of selecting tumors harboring p53 mutations that escape MDM2 control. In this study, we report a novel small-molecule inhibitor of the MDM2-p53 interaction, SAR405838 (MI-77301), that has been advanced into phase I clinical trials. SAR405838 binds to MDM2 with Ki = 0.88 nmol/L and has high specificity over other proteins. A cocrystal structure of the SAR405838:MDM2 complex shows that, in addition to mimicking three key p53 amino acid residues, the inhibitor captures additional interactions not observed in the p53-MDM2 complex and induces refolding of the short, unstructured MDM2 N-terminal region to achieve its high affinity. SAR405838 effectively activates wild-type p53 in vitro and in xenograft tumor tissue of leukemia and solid tumors, leading to p53-dependent cell-cycle arrest and/or apoptosis. At well-tolerated dose schedules, SAR405838 achieves either durable tumor regression or complete tumor growth inhibition in mouse xenograft models of SJSA-1 osteosarcoma, RS4;11 acute leukemia, LNCaP prostate cancer, and HCT-116 colon cancer. Remarkably, a single oral dose of SAR405838 is sufficient to achieve complete tumor regression in the SJSA-1 model. Mechanistically, robust transcriptional upregulation of PUMA induced by SAR405838 results in strong apoptosis in tumor tissue, leading to complete tumor regression. Our findings provide a preclinical basis upon which to evaluate SAR405838 as a therapeutic agent in patients whose tumors retain wild-type p53. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
244. Structural Basis for the Recognition of Peptide RJPXD33 by Acyltransferases in Lipid A Biosynthesis.
- Author
-
Jenkins, Ronald J., Heslip, Kyle A., Meagher, Jennifer L., Stuckey, Jeanne A., and Dotson, Garry D.
- Subjects
- *
PEPTIDES , *ACYLTRANSFERASES , *LIPIDS , *BIOSYNTHESIS , *PROTEIN binding , *BIOCHEMISTRY - Abstract
UDP-N-acetylglucosamine acyltransferase (LpxA) and UDP-3-O-(acyl)-glucosamine acyltransferase (LpxD) constitute the essential, early acyltransferases of lipid A biosynthesis. Recently, an antimicrobial peptide inhibitor, RJPXD33, was identified with dual affinity for LpxA and LpxD. To gain a fundamental understanding of the molecular basis of inhibitor binding, we determined the crystal structure of LpxA from Escherichia coli in complex with RJPXD33 at 1.9 Å resolutions. Our results suggest that the peptide binds in a unique modality that mimics (R)-β-hydroxyacyl pantetheine binding to LpxA and displays how the peptide binds exclusive of the native substrate, acyl-acyl carrier protein. Acyltransferase binding studies with photo-labile RJPXD33 probes and truncations of RJPXD33 validated the structure and provided fundamental insights for future design of small molecule inhibitors. Overlay of the LpxA-RJPXD33 structure with E. coli LpxD identified a complementary peptide binding pocket within LpxD and serves as a model for further biochemical characterization of RJPXD33 binding to LpxD. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
245. The Tail of KdsC CONFORMA TIONAL CHANGES CONTROL THE ACTIVITY OF A HALOA CID DEHALOGENASE SUPERFAMILY PHOSPHATASE.
- Author
-
Biswas, Tapan, Li Yi, Aggarwal, Parag, Wu, Jing, Rubin, John R., Stuckey, Jeanne A., Woodard, Ronald W., and Tsodikov, Oleg V.
- Subjects
- *
PHOSPHATASES , *GRAM-negative bacteria , *ENDOTOXINS , *DRUG resistance in microorganisms , *HALOACID dehalogenase , *ESCHERICHIA coli , *IMMUNOSPECIFICITY - Abstract
The phosphatase KdsC cleaves 3-deoxy-D-manno-octulosonate 8-phosphate to generate a molecule of inorganic phosphate and Kdo. Kdo is an essential component of the lipopolysaccharide envelope in Gram-negative bacteria. Because lipopolysaccharide is an important determinant of bacterial resistance and toxicity, KdsC is a potential target for novel antibacterial agents. KdsC belongs to the broad haloacid dehalogenase superfamily. In haloacid dehalogenase superfamily enzymes, substrate specificity and catalytic efficiency are generally dictated by a fold feature called the cap domain. It is therefore not clear why KdsC, which lacks a cap domain, is catalytically efficient and highly specific to 3-deoxy-D-manno-octulosonate 8-phosphate. Here, we present a set of seven structures of tetrameric Escherichia coli KdsC (ranging from 1.4 to 3.06 A in resolution) that model different intermediate states in its catalytic mechanism. A crystal structure of product-bound E. coli KdsC shows how the interface between adjacent mono- mers defines the active site pocket. Kdo is engaged in a network of polar and nonpolar interactions with residues at this interface, which explains substrate specificity. Furthermore, this structural and kinetic analysis strongly suggests that the binding of the flexible C-terminal region (tail) to the active site makes KdsC catalytically efficient by facilitating product release. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
246. Design and characterization of bivalent Smac-based peptides as antagonists of XIAP and development and validation of a fluorescence polarization assay for XIAP containing both BIR2 and BIR3 domains
- Author
-
Nikolovska-Coleska, Zaneta, Meagher, Jennifer L., Jiang, Sheng, Kawamoto, Steven A., Gao, Wei, Yi, Han, Qin, Dongguang, Roller, Peter P., Stuckey, Jeanne A., and Wang, Shaomeng
- Subjects
- *
APOPTOSIS , *CHROMOSOMES , *CELL nuclei , *GENETICS - Abstract
Abstract: XIAP (X-chromosome-linked inhibitor of apoptosis protein) is an inhibitor of apoptosis by binding to and inhibition of caspase-3 and caspase-7 through its BIR2 domain and caspase-9 through its BIR3 domain. Smac (second mitochondria-derived activator of caspases) protein is an endogenous antagonist of XIAP. Smac forms a dimer and concurrently binds both the BIR2 and BIR3 domains in XIAP, functioning as a highly efficient and potent cellular inhibitor of XIAP. In this article, we have designed and synthesized a bivalent Smac-based ligand (Smac-1) and its fluorescent labeled analogue (Smac-1F) and characterized their interaction with different constructs of XIAP. Our study demonstrates that bivalent Smac-based ligands bind concurrently to both the BIR2 and BIR3 domains of XIAP and are more than 500 times more potent than the corresponding monovalent Smac-based ligands. Bivalent Smac-based ligands also function as much more potent antagonists of XIAP than do the corresponding monovalent Smac-based ligands in cell-free functional assays. Using Smac-1F and XIAP containing both BIR2 and BIR3 domains, we also developed and validated a new fluorescence polarization-based assay. Hence, our designed bivalent Smac-based peptides mimic the mode of dimeric Smac protein in their interaction with XIAP containing both BIR2 and BIR3 domains and achieve extremely high potency in binding and functional assays. Our study provides new insights into the mode of action of bivalent Smac ligands targeting XIAP and a basis for the design and development of cell-permeable, bivalent Smac mimetics. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
247. Development and optimization of a binding assay for the XIAP BIR3 domain using fluorescence polarization
- Author
-
Nikolovska-Coleska, Zaneta, Wang, Renxiao, Fang, Xueliang, Pan, Hongguang, Tomita, York, Li, Peng, Roller, Peter P., Krajewski, Krzysztof, Saito, Naoyuki G., Stuckey, Jeanne A., and Wang, Shaomeng
- Subjects
- *
APOPTOSIS , *PROSPECTING , *CARRIER proteins , *CANCER cells - Abstract
The X-linked inhibitor of apoptosis protein (XIAP) is a potent cellular inhibitor of apoptosis. Designing small-molecule inhibitors that target the BIR3 domain of XIAP, where Smac/DIABLO (second mitochondria-derived activator of caspase/direct IAP-binding protein with low pI) and caspase-9 bind, is a promising strategy for inhibiting the antiapoptotic activity of XIAP and for overcoming apoptosis resistance of cancer cells mediated by XIAP. Herein, we report the development of a homogeneous high-throughput assay based on fluorescence polarization for measuring the binding affinities of small-molecule inhibitors to the BIR3 domain of XIAP. Among four fluorescent probes tested, a mutated N-terminal Smac peptide (AbuRPFK-(5-Fam)-NH2) showed the highest affinity (
Kd = 17.92 nM) and a large dynamic range (ΔmP=231±0.9 ), and was selected as the most suitable probe for the binding assay. The binding conditions (DMSO tolerance and stability) have been investigated. Under optimized conditions, aZ′ factor of 0.88 was achieved in a 96-well format for high-throughput screening. It was found that the popular Cheng–Prusoff equation is invalid for the calculation of the competitive inhibition constants (Ki values) for inhibitors in the FP-based competitive binding assay conditions, and accordingly, a new mathematical equation was developed, validated, and used to compute theKi values. An associated Web-based computer program was also developed for this task. Several known Smac peptides with high and low affinities have been evaluated under the assay conditions and the results obtained indicated that the FP-based competitive binding assay performs correctly as designed: it can quantitatively and accurately determine the binding affinities of Smac-based peptide inhibitors with a wide range of affinities, and is suitable for high-throughput screening of inhibitors binding to the XIAP BIR3 domain. [Copyright &y& Elsevier]- Published
- 2004
- Full Text
- View/download PDF
248. Crystal Structure of a Phosphoinositide Phosphatase, MTMR2: Insights into Myotubular Myopathy and Charcot-Marie-Tooth Syndrome
- Author
-
Begley, Michael J., Taylor, Gregory S., Kim, Soo-A, Veine, Donna M., Dixon, Jack E., and Stuckey, Jeanne A.
- Subjects
- *
PROTEINS , *LIPIDS , *GENETIC mutation - Abstract
Myotubularin-related proteins are a large subfamily of protein tyrosine phosphatases (PTPs) that dephosphorylate D3-phosphorylated inositol lipids. Mutations in members of the myotubularin family cause the human neuromuscular disorders myotubular myopathy and type 4B Charcot-Marie-Tooth syndrome. The crystal structure of a representative member of this family, MTMR2, reveals a phosphatase domain that is structurally unique among PTPs. A series of mutants are described that exhibit altered enzymatic activity and provide insight into the specificity of myotubularin phosphatases toward phosphoinositide substrates. The structure also reveals that the GRAM domain, found in myotubularin family phosphatases and predicted to occur in ∼180 proteins, is part of a larger motif with a pleckstrin homology (PH) domain fold. Finally, the MTMR2 structure will serve as a model for other members of the myotubularin family and provide a framework for understanding the mechanism whereby mutations in these proteins lead to disease. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
249. Discovery of ST2 Inhibitors: From Biomarker to Potential Drug Target in Graft-Versus-Host Disease.
- Author
-
Yang, Chao-Yie, Zhang, Jilu, Daguindau, Etienne, Bolten, Zachary, Chinnaswamy, Krishnapriya, Stuckey, Jeanne A., and Paczesny, Sophie
- Subjects
- *
BIOMARKERS , *DRUG target , *GRAFT versus host disease , *CLINICAL trials , *DRUG development , *THERAPEUTICS - Published
- 2016
- Full Text
- View/download PDF
250. Discovery of High-Affinity SMARCA2/4 Bromodomain Ligands and Development of Potent and Exceptionally Selective SMARCA2 PROTAC Degraders.
- Author
-
Leng L, Tu W, Yang L, Huang L, Wang M, Meagher JL, Chinnaswamy K, Allu SR, Rej RK, Tošović J, Harikrishnan L, Li Z, Sui Z, Stuckey JA, and Wang S
- Abstract
In the SWI/SNF chromatin-remodeling complex, the mutually exclusive catalytic ATPase subunits SMARCA2 and SMARCA4 proteins have a synthetic-lethal relationship. Selectively targeting SMARCA2 for degradation is a promising and new therapeutic strategy for human cancers harboring inactivated mutated SMARCA4. In this study, we report the design, synthesis, and biological evaluation of novel SMARCA2/4 ligands and our subsequent design of PROTAC degraders using high-affinity SMARCA ligands and VHL-1 ligands. Our efforts led to the discovery of high-affinity SMARCA2/4 bromodomain ligands and the development of a potent and selective SMARCA2 degrader and a highly potent SMARCA2/4 and PBRM1 degrader.
- Published
- 2025
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.