Network


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

Hotspot


Dive into the research topics where Daniel R. Dempsey is active.

Publication


Featured researches published by Daniel R. Dempsey.


FEBS Letters | 2014

Identification of an arylalkylamine N‐acyltransferase from Drosophila melanogaster that catalyzes the formation of long‐chain N‐acylserotonins

Daniel R. Dempsey; Kristen A. Jeffries; Ryan L. Anderson; Anne-Marie Carpenter; Santiago Rodriquez Opsina; David J. Merkler

Arylalkylamine N‐acyltransferase‐like 2 2 (AANATL2) from Drosophila melanogaster was expressed and shown to catalyze the formation of long‐chain N‐acylserotonins and N‐acydopamines. Subsequent identification of endogenous amounts of N‐acylserotonins and colocalization of these fatty acid amides and AANATL2 transcripts gives supporting evidence that AANATL2 has a role in the biosynthetic formation of these important cell signalling lipids.


FEBS Letters | 2014

Drosophila melanogaster as a model system to study long-chain fatty acid amide metabolism

Kristen A. Jeffries; Daniel R. Dempsey; Anita L. Behari; Ryan L. Anderson; David J. Merkler

Long‐chain fatty acid amides are cell‐signaling lipids identified in mammals and, recently, in invertebrates, as well. Many details regarding fatty acid amide metabolism remain unclear. Herein, we demonstrate that Drosophila melanogaster is an excellent model system for the study long‐chain fatty acid amide metabolism as we have quantified the endogenous levels of N‐acylglycines, N‐acyldopamines, N‐acylethanolamines, and primary fatty acid amides by LC/QTOF‐MS. Growth of D. melanogaster on media supplemented with [1‐13C]‐palmitate lead to a family of 13C‐palmitate‐labeled fatty acid amides in the fly heads. The [1‐13C]‐palmitate feeding studies provide insight into the biosynthesis of the fatty acid amides.


Molecular Cell | 2017

A Tunable Brake for HECT Ubiquitin Ligases

Zan Chen; Hanjie Jiang; Wei Xu; Xiaoguang Li; Daniel R. Dempsey; Xiangbin Zhang; Peter N. Devreotes; Cynthia Wolberger; L. Mario Amzel; Sandra B. Gabelli; Philip A. Cole

The HECT E3 ligases ubiquitinate numerous transcription factors and signaling molecules, and their activity must be tightly controlled to prevent cancer, immune disorders, and other diseases. In this study, we have found unexpectedly that peptide linkers tethering WW domains in several HECT family members are key regulatory elements of their catalytic activities. Biochemical, structural, and cellular analyses have revealed that the linkers can lock the HECT domain in an inactive conformation and block the proposed allosteric ubiquitin binding site. Such linker-mediated autoinhibition of the HECT domain can be relieved by linker post-translational modifications, but complete removal of the brake can induce hyperactive autoubiquitination and E3 self destruction. These results clarify the mechanisms of several HECT protein cancer associated mutations and provide a new framework for understanding how HECT ubiquitin ligases must be finely tuned to ensure normal cellular behavior.


Biochemistry | 2014

Mechanistic and Structural Analysis of Drosophila melanogaster Arylalkylamine N-Acetyltransferases

Daniel R. Dempsey; Kristen A. Jeffries; Jason D. Bond; Anne-Marie Carpenter; Santiago Rodriguez-Ospina; Leonid Breydo; K. Kenneth Caswell; David J. Merkler

Arylalkylamine N-acetyltransferase (AANAT) catalyzes the penultimate step in the biosynthesis of melatonin and other N-acetylarylalkylamides from the corresponding arylalkylamine and acetyl-CoA. The N-acetylation of arylalkylamines is a critical step in Drosophila melanogaster for the inactivation of the bioactive amines and the sclerotization of the cuticle. Two AANAT variants (AANATA and AANATB) have been identified in D. melanogaster, in which AANATA differs from AANATB by the truncation of 35 amino acids from the N-terminus. We have expressed and purified both D. melanogaster AANAT variants (AANATA and AANATB) in Escherichia coli and used the purified enzymes to demonstrate that this N-terminal truncation does not affect the activity of the enzyme. Subsequent characterization of the kinetic and chemical mechanism of AANATA identified an ordered sequential mechanism, with acetyl-CoA binding first, followed by tyramine. We used a combination of pH–activity profiling and site-directed mutagenesis to study prospective residues believed to function in AANATA catalysis. These data led to an assignment of Glu-47 as the general base in catalysis with an apparent pKa of 7.0. Using the data generated for the kinetic mechanism, structure–function relationships, pH–rate profiles, and site-directed mutagenesis, we propose a chemical mechanism for AANATA.


Nature Methods | 2016

Enzyme-catalyzed expressed protein ligation

Samuel Henager; Nam Chu; Zan Chen; David M. Bolduc; Daniel R. Dempsey; Yousang Hwang; James A. Wells; Philip A. Cole

Expressed protein ligation is a valuable method for protein semisynthesis that involves the reaction of recombinant protein C-terminal thioesters with N-terminal cysteine (N-Cys)-containing peptides, but the requirement of a Cys residue at the ligation junction can limit the utility of this method. Here we employ subtiligase variants to efficiently ligate Cys-free peptides to protein thioesters. Using this method, we have more accurately determined the effect of C-terminal phosphorylation on the tumor suppressor protein PTEN.


Biochemistry | 2015

Mechanistic and Structural Analysis of a Drosophila melanogaster Enzyme, Arylalkylamine N-Acetyltransferase Like 7, an Enzyme That Catalyzes the Formation of N-Acetylarylalkylamides and N-Acetylhistamine

Daniel R. Dempsey; Kristen A. Jeffries; Sumit Handa; Anne-Marie Carpenter; Santiago Rodriguez-Ospina; Leonid Breydo; David J. Merkler

Arylalkylamine N-acetyltransferase like 7 (AANATL7) catalyzes the formation of N-acetylarylalkylamides and N-acetylhistamine from acetyl-CoA and the corresponding amine substrate. AANATL7 is a member of the GNAT superfamily of >10000 GCN5-related N-acetyltransferases, many members being linked to important roles in both human metabolism and disease. Drosophila melanogaster utilizes the N-acetylation of biogenic amines for the inactivation of neurotransmitters, the biosynthesis of melatonin, and the sclerotization of the cuticle. We have expressed and purified D. melanogaster AANATL7 in Escherichia coli and used the purified enzyme to define the substrate specificity for acyl-CoA and amine substrates. Information about the substrate specificity provides insight into the potential contribution made by AANATL7 to fatty acid amide biosynthesis because D. melanogaster has emerged as an important model system contributing to our understanding of fatty acid amide metabolism. Characterization of the kinetic mechanism of AANATL7 identified an ordered sequential mechanism, with acetyl-CoA binding first followed by histamine to generate an AANATL7·acetyl-CoA·histamine ternary complex prior to catalysis. Successive pH-activity profiling and site-directed mutagenesis experiments identified two ionizable groups: one with a pKa of 7.1 that is assigned to Glu-26 as a general base and a second pKa of 9.5 that is assigned to the protonation of the thiolate of the coenzyme A product. Using the data generated herein, we propose a chemical mechanism for AANATL7 and define functions for other important amino acid residues involved in substrate binding and regulation of catalysis.


Bioorganic Chemistry | 2015

Mechanistic analysis of ghrelin-O-acyltransferase using substrate analogs.

Martin S. Taylor; Daniel R. Dempsey; Yousang Hwang; Zan Chen; Nam Chu; Jef D. Boeke; Philip A. Cole

Ghrelin-O-Acyltransferase (GOAT) is an 11-transmembrane integral membrane protein that octanoylates the metabolism-regulating peptide hormone ghrelin at Ser3 and may represent an attractive target for the treatment of type II diabetes and the metabolic syndrome. Protein octanoylation is unique to ghrelin in humans, and little is known about the mechanism of GOAT or of related protein-O-acyltransferases HHAT or PORC. In this study, we explored an in vitro microsomal ghrelin octanoylation assay to analyze its enzymologic features. Measurement of Km for 10-mer, 27-mer, and synthetic Tat-peptide-containing ghrelin substrates provided evidence for a role of charge interactions in substrate binding. Ghrelin substrates with amino-alanine in place of Ser3 demonstrated that GOAT can catalyze the formation of an octanoyl-amide bond at a similar rate compared with the natural reaction. A pH-rate comparison of these substrates revealed minimal differences in acyltransferase activity across pH 6.0-9.0, providing evidence that these reactions may be relatively insensitive to the basicity of the substrate nucleophile. The conserved His338 residue was required both for Ser3 and amino-Ala3 ghrelin substrates, suggesting that His338 may have a key catalytic role beyond that of a general base.


Protein Expression and Purification | 2014

Expression, purification, and characterization of mouse glycine N-acyltransferase in Escherichia coli

Daniel R. Dempsey; Jason D. Bond; Anne-Marie Carpenter; Santiago Rodriguez Ospina; David J. Merkler

Glycine N-acyltransferase (GLYAT) is a phase II metabolic detoxification enzyme for exogenous (xenobiotic) and endogenous carboxylic acids; consisting of fatty acids, benzoic acid, and salicylic acid. GLYAT catalyzes the formation of hippurate (N-benzoylglycine) from the corresponding glycine and benzoyl-CoA. Herein, we report the successful expression, purification, and characterization of recombinant mouse GLYAT (mGLYAT). A 34kDa mGLYAT protein was expressed in Escherichia coli and purified to homogeneity by nickel affinity chromatography to a final yield of 2.5mg/L culture. Characterization for both amino donors and amino acceptors were completed, with glycine serving as the best amino donor substrate, (kcat/Km)app=(5.2±0.20)×10(2)M(-1)s(-1), and benzoyl-CoA serving as the best the amino acceptor substrate, (kcat/Km)app=(4.5±0.27)×10(5)M(-1)s(-1). Our data demonstrate that mGLYAT will catalyzed the chain length specific (C2-C6) formation of N-acylglycines. The steady-state kinetic constants determined for recombinant mGLYAT for the substrates benzoyl-CoA and glycine, were shown to be consistent with other reported species (rat, human, bovine, ovine, and rhesus monkey). The successful recombinant expression and purification of mGLYAT can lead to solve unanswered questions associated with this enzyme, consisting of what is the chemical mechanism and what catalytic residues are essential for the how this phase II metabolic detoxification enzyme conjugates glycine to xenobiotic and endogenous carboxylic acids.


Insect Biochemistry and Molecular Biology | 2015

Probing the chemical mechanism and critical regulatory amino acid residues of Drosophila melanogaster arylalkylamine N-acyltransferase like 2.

Daniel R. Dempsey; Anne-Marie Carpenter; Santiago Rodriguez Ospina; David J. Merkler

Arylalkylamine N-acyltransferase like 2 (AANATL2) catalyzes the formation of N-acylarylalkylamides from the corresponding acyl-CoA and arylalkylamine. The N-acylation of biogenic amines in Drosophila melanogaster is a critical step for the inactivation of neurotransmitters, cuticle sclerotization, and melatonin biosynthesis. In addition, D. melanogaster has been used as a model system to evaluate the biosynthesis of fatty acid amides: a family of potent cell signaling lipids. We have previously showed that AANATL2 catalyzes the formation of N-acylarylakylamides, including long-chain N-acylserotonins and N-acyldopamines. Herein, we define the kinetic mechanism for AANATL2 as an ordered sequential mechanism with acetyl-CoA binding first followed by tyramine to generate the ternary complex prior to catalysis. Bell shaped kcat,app - acetyl-CoA and (kcat/Km)app - acetyl-CoA pH-rate profiles identified two apparent pKa,app values of ∼7.4 and ∼8.9 that are critical to catalysis, suggesting the AANATL2-catalyzed formation of N-acetyltyramine occurs through an acid/base chemical mechanism. Site-directed mutagenesis of a conserved glutamate that corresponds to the catalytic base for other D. melanogaster AANATL enzymes did not produce a substantial depression in the kcat,app value nor did it abolish the pKa,app value attributed to the general base in catalysis (pKa ∼7.4). These data suggest that AANATL2 catalyzes the formation of N-acylarylalkylamides using either different catalytic residues or a different chemical mechanism relative to other D. melanogaster AANATL enzymes. In addition, we constructed other site-directed mutants of AANATL2 to help define the role of targeted amino acids in substrate binding and/or enzyme catalysis.


Omega-3 Fatty Acids in Brain and Neurological Health | 2014

Mammalian Fatty Acid Amides of the Brain and CNS

Dominik P. Waluk; Matthew R. Battistini; Daniel R. Dempsey; Emma K. Farrell; Kristen A. Jeffries; Perry Mitchell; Lucas W. Hernandez; Joshua C. McBride; David J. Merkler; Mary C. Hunt

Fatty acid amides have emerged as an intriguing family of diverse, mammalian neuroactive lipids. Herein, we review the current state of our knowledge about the individual classes of fatty acid amides: which ones have been identified and characterized from mammals, their receptors, their functions, and the pathways for their biosynthesis and degradation. Much remains to be elucidated regarding this family of molecules and we hope that our review stimulates additional research in the fatty acid amide field. We also show that there are extensive metabolic connections between the different classess of fatty acid amides. Such metabolic connections hint at novel modes of regulation as one fatty acid amide is converted to another and suggests that the enzymes involved in fatty acid amide metabolism are not simply enzymes of biosynthesis or degradation, but also serve important regulatory functions.

Collaboration


Dive into the Daniel R. Dempsey's collaboration.

Top Co-Authors

Avatar

David J. Merkler

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zan Chen

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Philip A. Cole

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Nam Chu

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ryan L. Anderson

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sumit Handa

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Yousang Hwang

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge