Network


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

Hotspot


Dive into the research topics where Dewey G. McCafferty is active.

Publication


Featured researches published by Dewey G. McCafferty.


Biopolymers | 2010

Sortase transpeptidases: Insights into mechanism, substrate specificity, and inhibition†

Kathleen W. Clancy; Jeffrey A. Melvin; Dewey G. McCafferty

Gram-positive bacteria pose a serious healthcare threat. The growing antibiotic resistance epidemic creates a dire need for new antibiotic targets. The sortase family of enzymes is a promising target for antimicrobial therapy. This review covers the current knowledge of the mechanism, substrate specificity, and inhibitory studies of the Gram-positive bacterial [corrected] enzyme sortase.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Facile synthesis of site-specifically acetylated and methylated histone proteins: Reagents for evaluation of the histone code hypothesis

Shu He; David R. Bauman; Jamaine S. Davis; Alejandra Loyola; Kenichi Nishioka; Jennifer L. Gronlund; Danny Reinberg; Fanyu Meng; Neil L. Kelleher; Dewey G. McCafferty

The functional capacity of genetically encoded histone proteins can be powerfully expanded by posttranslational modification. A growing body of biochemical and genetic evidence clearly links the unique combinatorial patterning of side chain acetylation, methylation, and phosphorylation mainly within the highly conserved N termini of histones H2A, H2B, H3, and H4 with the regulation of gene expression and chromatin assembly and remodeling, in effect constituting a “histone code” for epigenetic signaling. Deconvoluting this code has proved challenging given the inherent posttranslational heterogeneity of histone proteins isolated from biological sources. Here we describe the application of native chemical ligation to the preparation of full-length histone proteins containing site-specific acetylation and methylation modifications. Peptide thioesters corresponding to histone N termini were prepared by solid phase peptide synthesis using an acid labile Boc/HF assembly strategy, then subsequently ligated to recombinantly produced histone C-terminal globular domains containing an engineered N-terminal cysteine residue. The ligation site is then rendered traceless by hydrogenolytic desulfurization, generating a native histone protein sequence. Synthetic histones generated by this method are fully functional, as evidenced by their self-assembly into a higher order H3/H4 heterotetramer, their deposition into nucleosomes by human ISWI-containing (Imitation of Switch) factor RSF (Remodeling and Spacing Factor), and by enzymatic modification by human Sirt1 deacetylase and G9a methyltransferase. Site-specifically modified histone proteins generated by this method will prove invaluable as novel reagents for the evaluation of the histone code hypothesis and analysis of epigenetic signaling mechanisms.


Infection and Immunity | 2009

Critical Role of NOD2 in Regulating the Immune Response to Staphylococcus aureus

Hitesh Deshmukh; James B. Hamburger; Sun Hee Ahn; Dewey G. McCafferty; Suxiao R. Yang; Vance G. Fowler

ABSTRACT NOD2 (the nucleotide-binding oligomerization domain containing protein 2) is known to be involved in host recognition of bacteria, although its role in the host response to Staphylococcus aureus infection is unknown. NOD2-deficient (Nod2−/−) mice and wild-type (WT) littermate controls were injected intraperitoneally with S. aureus suspension (107 bacteria/g of body weight), and their survival was monitored. Cultured bone marrow-derived neutrophils were harvested from Nod2−/− and WT mice and tested for cytokine production and phagocytosis. Compared to WT mice, Nod2−/− mice were significantly more susceptible to S. aureus infection (median survival of 1.5 days versus >5 days; P = 0.003) and had a significantly higher bacterial tissue burden. Cultured bone marrow-derived neutrophils from Nod2−/− and WT mice had similar levels of peritoneal neutrophil recruitment and intracellular killing, but bone marrow-derived neutrophils from Nod2−/− mice had significantly reduced ability to internalize fluorescein-labeled S. aureus. Nod2−/− mice had significantly higher levels of Th1-derived cytokines in serum (tumor necrosis factor alpha, gamma interferon, and interleukin-2 [IL-2]) compared to WT mice, whereas the levels of Th2-derived cytokines (IL-1β, IL-4, IL-6, and IL-10) were similar in Nod2−/− and WT mice. Thus, mice deficient in NOD2 are more susceptible to S. aureus. Increased susceptibility is due in part to defective neutrophil phagocytosis, elevated serum levels of Th1 cytokines, and a higher bacterial tissue burden.


Bioorganic & Medicinal Chemistry Letters | 2008

Facile synthesis of substituted trans-2-arylcyclopropylamine inhibitors of the human histone demethylase LSD1 and monoamine oxidases A and B.

David M. Gooden; Dawn M. Z. Schmidt; Julie A. Pollock; Ami M. Kabadi; Dewey G. McCafferty

A facile synthetic route to substituted trans-2-arylcyclopropylamines was developed to provide access to mechanism-based inhibitors of the human flavoenzyme oxidase lysine-specific histone demethylase LSD1 and related enzyme family members such as monoamine oxidases A and B.


Tetrahedron | 1995

Synthesis of redox derivatives of lysine and their use in solid-phase synthesis of a light-harvesting peptide

Dewey G. McCafferty; Barney M. Bishop; Craig G. Wall; Solon G. Hughes; Sandra L. Mecklenberg; Thomas J. Meyer; Bruce W. Erickson

Redox-active amino acids were synthesized for incorporation into peptide assemblies to study photoinitiated electron or energy transfer. 4′-Methyl-2,2′-bipyridine-4-carboxylic acid was obtained in 72% yield by consecutive SeO2 and Ag2O oxidation without isolation of intermediates. The side chain e-amino group of Boc-l-lysine methyl ester or γ-carboxyl group of Boc-l-glutamic acid α-methyl ester was coupled to a redox moiety (transition-metal chromophore, electron donor, electron acceptor, metal ligand, or triplet-energy transmitter) using 4-(dimethylamino)pyridine, (1-benzotriazoleoxy)tris(dimethylamino)phosphonium hexafluorophosphate, N-methylmorpholine, and 1-hydroxybenzotriazole. Use of one equivalent of 4-(dimethylamino)pyridine provided the amide coupling product in 80–97% isolated yield. Selective hydrolysis of the methyl esters with lithium hydroxide provided the redox Boc-amino acids in 70–98% yield. These redox modules are suitable for solid-phase assembly of light-harvesting peptides, as illustrated by the synthesis of the partially α-helical 11-residue redox triad that contains a phenothiazine electron donor, a ruthenium(II)tris(bipyridine) chromophore, and an anthraquinone electron acceptor. Upon laser excitation at 420 nm, the peptide triad underwent photoinduced electron transfer to create a charge-separated state with a lifetime of 53 ns and decayed with a first-order rate constant of 1.9 × 108 s−1.


Current Opinion in Chemical Biology | 1999

Synergy and duality in peptide antibiotic mechanisms

Dewey G. McCafferty; Predrag Cudic; Michael K. Yu; Douglas C. Behenna; Ryan G. Kruger

The molecular mechanisms by which peptide antibiotics disrupt bacterial DNA synthesis, protein biosynthesis, cell wall biosynthesis, and membrane integrity are diverse, yet historically have been understood to follow a theme of one antibiotic, one inhibitory mechanism. In the past year, mechanistic and structural studies have shown a rich diversity in peptide antibiotic mechanism. Novel secondary targeting mechanisms for peptide antibiotics have recently been discovered, and the mechanisms of peptide antibiotics involved in synergistic relationships with antibiotics and proteins have been more clearly defined. In apparent response to selective pressures, antibiotic-producing organisms have elegantly integrated multiple functions and cooperative interactions into peptide antibiotic design for the purpose of improving antimicrobial success.


Journal of Biological Chemistry | 2009

Crystal Structure of Streptococcus pyogenes Sortase A: Implications for Sortase mechanism

Paul R. Race; Matthew L. Bentley; Jeff A. Melvin; Allister Crow; Richard K. Hughes; Wendy Smith; Richard B. Sessions; Michael A. Kehoe; Dewey G. McCafferty; Mark J. Banfield

Sortases are a family of Gram-positive bacterial transpeptidases that anchor secreted proteins to bacterial cell surfaces. These include many proteins that play critical roles in the virulence of Gram-positive bacterial pathogens such that sortases are attractive targets for development of novel antimicrobial agents. All Gram-positive pathogens express a “housekeeping” sortase that recognizes the majority of secreted proteins containing an LPXTG wall-sorting motif and covalently attaches these to bacterial cell wall peptidoglycan. Many Gram-positive pathogens also express additional sortases that link a small number of proteins, often with variant wall-sorting motifs, to either other surface proteins or peptidoglycan. To better understand the mechanisms of catalysis and substrate recognition by the housekeeping sortase produced by the important human pathogen Streptococcus pyogenes, the crystal structure of this protein has been solved and its transpeptidase activity established in vitro. The structure reveals a novel arrangement of key catalytic residues in the active site of a sortase, the first that is consistent with kinetic analysis. The structure also provides a complete description of residue positions surrounding the active site, overcoming the limitation of localized disorder in previous structures of sortase A-type proteins. Modification of the active site Cys through oxidation to its sulfenic acid form or by an alkylating reagent supports a role for a reactive thiol/thiolate in the catalytic mechanism. These new insights into sortase structure and function could have important consequences for inhibitor design.


Chemistry & Biology | 1998

Homologs of the vancomycin resistance D-Ala-D-Ala dipeptidase VanX in Streptomyces toyocaensis, Escherichia coli and Synechocystis: attributes of catalytic efficiency, stereoselectivity and regulation with implications for function.

Ivan A.D. Lessard; Steve D. Pratt; Dewey G. McCafferty; Dirksen E. Bussiere; Charles Hutchins; Barry L. Wanner; Leo Katz; Christopher T. Walsh

BACKGROUND Vancomycin-resistant enterococci are pathogenic bacteria that have altered cell-wall peptidoglycan termini (D-alanyl-D-lactate [D-Ala-D-lactate] instead of D-alanyl-D-alanine [D-Ala-D-Ala]), which results in a 1000-fold decreased affinity for binding vancomycin. The metallodipeptidase VanX (EntVanX) is key enzyme in antibiotic resistance as it reduces the cellular pool of the D-Ala-D-Ala dipeptide. RESULTS A bacterial genome search revealed vanX homologs in Streptomyces toyocaensis (StoVanX), Escherichia coli (EcoVanX), and Synechocystis sp. strain PCC6803 (SynVanX). Here, the D,D-dipeptidase catalytic activity of all three VanX homologs is validated, and the catalytic efficiencies and diastereoselectivity ratios for dipeptide cleavage are reported. The ecovanX gene is shown to have an RpoS (sigma(s))-dependent promoter typical of genes turned on in stationary phase. Expression of ecovanX and an associated cluster of dipeptide permease genes permitted growth of E. coli using D-Ala-D-Ala as the sole carbon source. CONCLUSIONS The key residues of the EntVanX active site are strongly conserved in the VanX homologs, suggesting their active-site topologies are similar. StoVanX is a highly efficient D-Ala-D-Ala dipeptidase; its gene is located in a vanHAX operon, consistent with a vancomycin-immunity function. StoVanX is a potential source for the VanX found in gram-positive enterococci. The catalytic efficiencies of D-Ala-D-Ala hydrolysis for EcoVanX and SynVanX are 25-fold lower than for EntVanX, suggesting they have a role in cell-wall turnover. Clustered with the ecovanX gene is a putative dipeptide permease system that imports D-Ala-D-Ala into the cell. The combined action of EcoVanX and the permease could permit the use of D-Ala-D-Ala as a bacterial energy source under starvation conditions.


Journal of Biological Chemistry | 2008

Mutagenesis Studies of Substrate Recognition and Catalysis in the Sortase A Transpeptidase from Staphylococcus aureus

Matthew L. Bentley; Erin C. Lamb; Dewey G. McCafferty

The Staphylococcus aureus transpeptidase sortase A (SrtA) is responsible for anchoring a range of virulence- and colonization-associated proteins to the cell wall. SrtA recognizes substrates that contain a C-terminal LPXTG motif. This sequence is cleaved following the threonine, and an amide bond is formed between the threonine and the pentaglycine cross-bridge of branched lipid II. Previous studies have implicated the β6/β7 loop region of SrtA in LPXTG recognition but have not systematically characterized this domain. To better understand the individual roles of the residues within this loop, we performed alanine-scanning mutagenesis. Val-168 and Leu-169 were found to be important for substrate recognition, and Glu-171 was also found to be important, consistent with its hypothesized role as a Ca2+-binding residue. Gly-167 and Asp-170 were dispensable for catalysis, as was Gln-172. The role of Arg-197 in SrtA has been the subject of much debate. To explore its role in catalysis, we used native chemical ligation to generate semi-synthetic SrtA in which we replaced Arg-197 with citrulline, a non-ionizable analog. This change resulted in a decrease of <3-fold in kcat/Km, indicating that Arg-197 utilizes a hydrogen bond, rather than an electrostatic interaction. Our results are consistent with a model for LPXTG recognition wherein the Leu-Pro sequence is recognized primarily by hydrophobic contacts with SrtA Val-168 and Leu-169, as well as a hydrogen bond from Arg-197. This model contradicts the previously proposed mechanism of binding predicted by the x-ray crystal structure of SrtA.


Journal of Biological Chemistry | 2007

Engineering the substrate specificity of staphylococcus aureus sortase A: The β6/β7 loop from SrtB confers NPQTN recognition to SrtA

Matthew L. Bentley; Helena Gaweska; Joseph M. Kielec; Dewey G. McCafferty

The Staphylococcus aureus transpeptidase Sortase A (SrtA) anchors virulence and colonization-associated surface proteins to the cell wall. SrtA selectively recognizes a C-terminal LPXTG motif, whereas the related transpeptidase Sortase B (SrtB) recognizes a C-terminal NPQTN motif. In both enzymes, cleavage occurs after the conserved threonine, followed by amide bond formation between threonine and the pentaglycine cross-bridge of cell wall peptidoglycan. Genetic and biochemical studies strongly suggest that SrtA and SrtB exhibit exquisite specificity for their recognition motifs. To better understand the origins of substrate specificity within these two isoforms, we used sequence and structural analysis to predict residues and domains likely to be involved in conferring substrate specificity. Mutational analyses and domain swapping experiments were conducted to test their function in substrate recognition and specificity. Marked changes in the specificity profile of SrtA were obtained by replacing the β6/β7 loop in SrtA with the corresponding domain from SrtB. The chimeric β6/β7 loop swap enzyme (SrtLS) conferred the ability to acylate NPQTN-containing substrates, with a \batchmode \documentclass[fleqn,10pt,legalpaper]{article} \usepackage{amssymb} \usepackage{amsfonts} \usepackage{amsmath} \pagestyle{empty} \begin{document} \(k_{\mathrm{cat}}{/}K_{m}^{\mathrm{app}}\) \end{document} of 0.0062 ± 0.003 m-1 s-1. This enzyme was unable to perform the transpeptidation stage of the reaction, suggesting that additional domains are required for transpeptidation to occur. The overall catalytic specificity profile (\batchmode \documentclass[fleqn,10pt,legalpaper]{article} \usepackage{amssymb} \usepackage{amsfonts} \usepackage{amsmath} \pagestyle{empty} \begin{document} \(k_{\mathrm{cat}}{/}K_{m}^{\mathrm{app}}(\mathrm{NPQTN}){/}k_{\mathrm{cat}}{/}K_{m}^{\mathrm{app}}(\mathrm{LPETG})\) \end{document}) of SrtLS was altered 700,000-fold from SrtA. These results indicate that the β6/β7 loop is an important site for substrate recognition in sortases.

Collaboration


Dive into the Dewey G. McCafferty's collaboration.

Top Co-Authors

Avatar

Ryan G. Kruger

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bruce W. Erickson

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Brenda A. Frankel

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Predrag Cudic

Florida Atlantic University

View shared research outputs
Top Co-Authors

Avatar

Thomas J. Meyer

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Craig G. Wall

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick Dostal

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge