Network


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

Hotspot


Dive into the research topics where Frank Zucker is active.

Publication


Featured researches published by Frank Zucker.


Antimicrobial Agents and Chemotherapy | 2011

Selective Inhibitors of Methionyl-tRNA Synthetase Have Potent Activity against Trypanosoma brucei Infection in Mice

Sayaka Shibata; J. Robert Gillespie; Angela Kelley; Alberto J. Napuli; Zhongsheng Zhang; Kuzma V. Kovzun; Ranae Pefley; Jocelyn Lam; Frank Zucker; Wesley C. Van Voorhis; Ethan A. Merritt; Wim G. J. Hol; Christophe L. M. J. Verlinde; Erkang Fan; Frederick S. Buckner

ABSTRACT Human African trypanosomiasis continues to be an important public health threat in extensive regions of sub-Saharan Africa. Treatment options for infected patients are unsatisfactory due to toxicity, difficult administration regimes, and poor efficacy of available drugs. The aminoacyl-tRNA synthetases were selected as attractive drug targets due to their essential roles in protein synthesis and cell survival. Comparative sequence analysis disclosed differences between the trypanosome and mammalian methionyl-tRNA synthetases (MetRSs) that suggested opportunities for selective inhibition using drug-like molecules. Experiments using RNA interference on the single MetRS of Trypanosoma brucei demonstrated that this gene product was essential for normal cell growth. Small molecules (diaryl diamines) similar to those shown to have potent activity on prokaryotic MetRS enzymes were synthesized and observed to have inhibitory activity on the T. brucei MetRS (50% inhibitory concentration, <50 nM) and on bloodstream forms of T. brucei cultures (50% effective concentration, as low as 4 nM). Twenty-one compounds had a close correlation between enzyme binding/inhibition and T. brucei growth inhibition, indicating that they were likely to be acting on the intended target. The compounds had minimal effects on mammalian cell growth at 20 μM, demonstrating a wide therapeutic index. The most potent compound was tested in the murine model of trypanosomiasis and demonstrated profound parasite suppression and delayed mortality. A homology model of the T. brucei MetRS based on other MetRS structures was used to model binding of the lead diaryl diamine compounds. Future studies will focus on improving the pharmacological properties of the MetRS inhibitors.


Journal of Medicinal Chemistry | 2012

Urea-based inhibitors of Trypanosoma brucei methionyl-tRNA synthetase: selectivity and in vivo characterization.

Sayaka Shibata; J. Robert Gillespie; Ranae M. Ranade; Cho Yeow Koh; Jessica E. Kim; Joy U. Laydbak; Frank Zucker; Wim G. J. Hol; Christophe L. M. J. Verlinde; Frederick S. Buckner; Erkang Fan

Urea-based methionyl-tRNA synthetase inhibitors were designed, synthesized, and evaluated for their potential toward treating human African trypanosomiasis (HAT). With the aid of a homology model and a structure-activity-relationship approach, low nM inhibitors were discovered that show high selectivity toward the parasite enzyme over the closest human homologue. These compounds inhibit parasite growth with EC(50) values as low as 0.15 μM while having low toxicity to mammalian cells. Two compounds (2 and 26) showed excellent membrane permeation in the MDR1-MDCKII model and encouraging oral pharmacokinetic properties in mice. Compound 2 was confirmed to enter the CNS in mice. Compound 26 had modest suppressive activity against Trpanosoma brucei rhodesiense in the mouse model, suggesting that more potent analogues or compounds with higher exposures need to be developed. The urea-based inhibitors are thus a promising starting point for further optimization toward the discovery of orally available and CNS active drugs to treat HAT.


Acta Crystallographica Section D-biological Crystallography | 2010

Validation of crystallographic models containing TLS or other descriptions of anisotropy.

Frank Zucker; P. Christoph Champ; Ethan A. Merritt

The use of TLS (translation/libration/screw) models to describe anisotropic displacement of atoms within a protein crystal structure has become increasingly common. These models may be used purely as an improved methodology for crystallographic refinement or as the basis for analyzing inter-domain and other large-scale motions implied by the crystal structure. In either case it is desirable to validate that the crystallographic model, including the TLS description of anisotropy, conforms to our best understanding of protein structures and their modes of flexibility. A set of validation tests has been implemented that can be integrated into ongoing crystallographic refinement or run afterwards to evaluate a previously refined structure. In either case validation can serve to increase confidence that the model is correct, to highlight aspects of the model that may be improved or to strengthen the evidence supporting specific modes of flexibility inferred from the refined TLS model. Automated validation checks have been added to the PARVATI and TLSMD web servers and incorporated into the CCP4i user interface.


Journal of Molecular Biology | 2010

Crystal Structures of Trypanosomal Histidyl-tRNA Synthetase Illuminate Differences between Eukaryotic and Prokaryotic Homologs ☆

Ethan A. Merritt; Tracy L. Arakaki; J. Robert Gillespie; Eric T. Larson; Angela Kelley; Natascha Mueller; Alberto J. Napuli; Jessica E. Kim; Li Zhang; Christophe L. M. J. Verlinde; Erkang Fan; Frank Zucker; Frederick S. Buckner; Wesley C. Van Voorhis; Wim G. J. Hol

Crystal structures of histidyl-tRNA synthetase (HisRS) from the eukaryotic parasites Trypanosoma brucei and Trypanosoma cruzi provide a first structural view of a eukaryotic form of this enzyme and reveal differences from bacterial homologs. HisRSs in general contain an extra domain inserted between conserved motifs 2 and 3 of the Class II aminoacyl-tRNA synthetase catalytic core. The current structures show that the three-dimensional topology of this domain is very different in bacterial and archaeal/eukaryotic forms of the enzyme. Comparison of apo and histidine-bound trypanosomal structures indicates substantial active-site rearrangement upon histidine binding but relatively little subsequent rearrangement after reaction of histidine with ATP to form the enzymes first reaction product, histidyladenylate. The specific residues involved in forming the binding pocket for the adenine moiety differ substantially both from the previously characterized binding site in bacterial structures and from the homologous residues in human HisRSs. The essentiality of the single HisRS gene in T. brucei is shown by a severe depression of parasite growth rate that results from even partial suppression of expression by RNA interference.


Journal of Molecular Biology | 2011

The Double-Length Tyrosyl-tRNA Synthetase from the Eukaryote Leishmania major Forms an Intrinsically Asymmetric Pseudo-Dimer.

Eric T. Larson; Jessica E. Kim; Lisa J. Castaneda; Alberto J. Napuli; Zhongsheng Zhang; Erkang Fan; Frank Zucker; Christophe L. M. J. Verlinde; Frederick S. Buckner; Wesley C. Van Voorhis; Wim G. J. Hol; Ethan A. Merritt

The single tyrosyl-tRNA synthetase (TyrRS) gene in trypanosomatid genomes codes for a protein that is twice the length of TyrRS from virtually all other organisms. Each half of the double-length TyrRS contains a catalytic domain and an anticodon-binding domain; however, the two halves retain only 17% sequence identity to each other. The structural and functional consequences of this duplication and divergence are unclear. TyrRS normally forms a homodimer in which the active site of one monomer pairs with the anticodon-binding domain from the other. However, crystal structures of Leishmania major TyrRS show that, instead, the two halves of a single molecule form a pseudo-dimer resembling the canonical TyrRS dimer. Curiously, the C-terminal copy of the catalytic domain has lost the catalytically important HIGH and KMSKS motifs characteristic of class I aminoacyl-tRNA synthetases. Thus, the pseudo-dimer contains only one functional active site (contributed by the N-terminal half) and only one functional anticodon recognition site (contributed by the C-terminal half). Despite biochemical evidence for negative cooperativity between the two active sites of the usual TyrRS homodimer, previous structures have captured a crystallographically-imposed symmetric state. As the L. major TyrRS pseudo-dimer is inherently asymmetric, conformational variations observed near the active site may be relevant to understanding how the state of a single active site is communicated across the dimer interface. Furthermore, substantial differences between trypanosomal TyrRS and human homologs are promising for the design of inhibitors that selectively target the parasite enzyme.


Current Topics in Medicinal Chemistry | 2009

Fragment-based cocktail crystallography by the medical structural genomics of pathogenic protozoa consortium.

Christophe L. M. J. Verlinde; Erkang Fan; Sayaka Shibata; Zhongsheng Zhang; Zhihua Sun; Wei Deng; Jennifer M. Ross; Jessica E. Kim; Liren Xiao; Tracy L. Arakaki; Jürgen Bosch; Jonathan M. Caruthers; Eric T. Larson; Isolde LeTrong; Alberto J. Napuli; Angela Kelley; Natasha Mueller; Frank Zucker; Wesley C. Van Voorhis; Frederick S. Buckner; Ethan A. Merritt; Wim G. J. Hol

The history of fragment-based drug discovery, with an emphasis on crystallographic methods, is sketched, illuminating various contributions, including our own, which preceded the industrial development of the method. Subsequently, the creation of the BMSC fragment cocktails library is described. The BMSC collection currently comprises 68 cocktails of 10 compounds that are shape-wise diverse. The utility of these cocktails for initiating lead discovery in structure-based drug design has been explored by soaking numerous protein crystals obtained by our MSGPP (Medical Structural Genomics of Pathogenic Protozoa) consortium. Details of the fragment selection and cocktail design procedures, as well as examples of the successes obtained are given. The BMSC Fragment Cocktail recipes are available free of charge and are in use in over 20 academic labs.


Proteins | 2005

Crystal structure of glyceraldehyde‐3‐phosphate dehydrogenase from Plasmodium falciparum at 2.25 Å resolution reveals intriguing extra electron density in the active site

Mark A. Robien; Jürgen Bosch; Frederick S. Buckner; Wesley C. Van Voorhis; Elizabeth A. Worthey; Peter J. Myler; Christopher Mehlin; Erica Boni; Oleksandr Kalyuzhniy; Lori Anderson; Angela Lauricella; Stacy Gulde; Joseph R. Luft; George T. DeTitta; Jonathan M. Caruthers; Keith O. Hodgson; Michael Soltis; Frank Zucker; Christophe L. M. J. Verlinde; Ethan A. Merritt; Lori W. Schoenfeld; Wim G. J. Hol

The crystal structure of D‐glyceraldehyde‐3‐phosphate dehydrogenase (PfGAPDH) from the major malaria parasite Plasmodium falciparum is solved at 2.25 Å resolution. The structure of PfGAPDH is of interest due to the dependence of the malaria parasite in infected human erythrocytes on the glycolytic pathway for its energy generation. Recent evidence suggests that PfGAPDH may also be required for other critical activities such as apical complex formation. The cofactor NAD+ is bound to all four subunits of the tetrameric enzyme displaying excellent electron densities. In addition, in all four subunits a completely unexpected large island of extra electron density in the active site is observed, approaching closely the nicotinamide ribose of the NAD+. This density is most likely the protease inhibitor AEBSF, found in maps from two different crystals. This putative AEBSF molecule is positioned in a crucial location and hence our structure, with expected and unexpected ligands bound, can be of assistance in lead development and design of novel antimalarials. Proteins 2006.


Protein Science | 2005

Crystal structures and proposed structural/functional classification of three protozoan proteins from the isochorismatase superfamily.

Jonathan M. Caruthers; Frank Zucker; Elizabeth A. Worthey; Peter J. Myler; Fred Buckner; Wes Van Voorhuis; Chris Mehlin; Erica Boni; Tiffany Feist; Joseph R. Luft; Stacey Gulde; Angela Lauricella; Oleksandr Kaluzhniy; Lori Anderson; Isolde Le Trong; Margaret A. Holmes; Thomas Earnest; Michael Soltis; Keith O. Hodgson; Wim G. J. Hol; Ethan A. Merritt

We have determined the crystal structures of three homologous proteins from the pathogenic protozoans Leishmania donovani, Leishmania major, and Trypanosoma cruzi. We propose that these proteins represent a new subfamily within the isochorismatase superfamily (CDD classification cd004310). Their overall fold and key active site residues are structurally homologous both to the biochemically well‐characterized N‐carbamoylsarcosine‐amidohydrolase, a cysteine hydrolase, and to the phenazine biosynthesis protein PHZD (isochorismase), an aspartyl hydrolase. All three proteins are annotated as mitochondrial‐associated ribonuclease Mar1, based on a previous characterization of the homologous protein from L. tarentolae. This would constitute a new enzymatic activity for this structural superfamily, but this is not strongly supported by the observed structures. In these protozoan proteins, the extended active site is formed by inter‐subunit association within a tetramer, which implies a distinct evolutionary history and substrate specificity from the previously characterized members of the isochorismatase superfamily. The characterization of the active site is supported crystallographically by the presence of an unidentified ligand bound at the active site cysteine of the T. cruzi structure.


Journal of Molecular Biology | 2010

The crystal structure and activity of a putative trypanosomal nucleoside phosphorylase reveal it to be a homodimeric uridine phosphorylase.

Eric T. Larson; Devaraja G. Mudeppa; J. Robert Gillespie; Natascha Mueller; Alberto J. Napuli; Jennifer A. Arif; Jenni Ross; Tracy L. Arakaki; Angela Lauricella; George T. DeTitta; Joseph R. Luft; Frank Zucker; Christophe L. M. J. Verlinde; Erkang Fan; Wesley C. Van Voorhis; Frederick S. Buckner; Pradipsinh K. Rathod; Wim G. J. Hol; Ethan A. Merritt

Purine nucleoside phosphorylases (PNPs) and uridine phosphorylases (UPs) are closely related enzymes involved in purine and pyrimidine salvage, respectively, which catalyze the removal of the ribosyl moiety from nucleosides so that the nucleotide base may be recycled. Parasitic protozoa generally are incapable of de novo purine biosynthesis; hence, the purine salvage pathway is of potential therapeutic interest. Information about pyrimidine biosynthesis in these organisms is much more limited. Though all seem to carry at least a subset of enzymes from each pathway, the dependency on de novo pyrimidine synthesis versus salvage varies from organism to organism and even from one growth stage to another. We have structurally and biochemically characterized a putative nucleoside phosphorylase (NP) from the pathogenic protozoan Trypanosoma brucei and find that it is a homodimeric UP. This is the first characterization of a UP from a trypanosomal source despite this activity being observed decades ago. Although this gene was broadly annotated as a putative NP, it was widely inferred to be a purine nucleoside phosphorylase. Our characterization of this trypanosomal enzyme shows that it is possible to distinguish between PNP and UP activity at the sequence level based on the absence or presence of a characteristic UP-specificity insert. We suggest that this recognizable feature may aid in proper annotation of the substrate specificity of enzymes in the NP family.


Proteins | 2005

Structure of a ribulose 5‐phosphate 3‐epimerase from Plasmodium falciparum

Jonathan M. Caruthers; Jürgen Bosch; Frederick S. Buckner; W. C. Van Voorhis; Peter J. Myler; Elizabeth A. Worthey; Christopher Mehlin; Erica Boni; George T. DeTitta; Joseph R. Luft; Angela Lauricella; Oleksandr Kalyuzhniy; Lori Anderson; Frank Zucker; Michael Soltis; Wim G. J. Hol

The crystal structure of Pfal009167AAA, a putative ribulose 5‐phosphate 3‐epimerase (PfalRPE) from Plasmodium falciparum, has been determined to 2 Å resolution. RPE represents an exciting potential drug target for developing antimalarials because it is involved in the shikimate and the pentose phosphate pathways. The structure is a classic TIM‐barrel fold. A coordinated Zn ion and a bound sulfate ion in the active site of the enzyme allow for a greater understanding of the mechanism of action of this enzyme. This structure is solved in the framework of the Structural Genomics of Pathogenic Protozoa (SGPP) consortium. Proteins 2006.

Collaboration


Dive into the Frank Zucker's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wim G. J. Hol

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erkang Fan

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph R. Luft

Hauptman-Woodward Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge